cancer vaccines - worcester polytechnic institute

117
1 CANCER VACCINES An Interactive Qualifying Project Report Submitted to the Faculty of WORCESTER POLYTECHNIC INSTITUTE In partial fulfillment of the requirements for the Degree of Bachelor of Science By: ____________________ ____________________ ____________________ Leila Camplese Johan Girgenrath Talal Hamza IQP-43-DSA-5218 IQP-43-DSA-7424 IQP-43-DSA-5103 ____________________ ____________________ Michaela Hunter Cole Royer IQP-43-DSA-7267 IQP-43-DSA-3408 August 13, 2018 APPROVED: _________________________ Prof. David S. Adams, PhD WPI Project Advisor This report represents the work of WPI undergraduate students submitted to the faculty as evidence of completion of a degree requirement. WPI routinely publishes these reports on its website without editorial or peer review. For more information about the projects program at WPI, please see http://www.wpi.edu/academics/ugradstudies/project-learning.html

Upload: others

Post on 16-Oct-2021

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CANCER VACCINES - Worcester Polytechnic Institute

1

CANCER VACCINES

An Interactive Qualifying Project Report

Submitted to the Faculty of

WORCESTER POLYTECHNIC INSTITUTE

In partial fulfillment of the requirements for the

Degree of Bachelor of Science

By:

____________________ ____________________ ____________________

Leila Camplese Johan Girgenrath Talal Hamza

IQP-43-DSA-5218 IQP-43-DSA-7424 IQP-43-DSA-5103

____________________ ____________________

Michaela Hunter Cole Royer

IQP-43-DSA-7267 IQP-43-DSA-3408

August 13, 2018

APPROVED:

_________________________

Prof. David S. Adams, PhD

WPI Project Advisor

This report represents the work of WPI undergraduate students submitted to the faculty as evidence of

completion of a degree requirement. WPI routinely publishes these reports on its website without

editorial or peer review. For more information about the projects program at WPI, please see

http://www.wpi.edu/academics/ugradstudies/project-learning.html

Page 2: CANCER VACCINES - Worcester Polytechnic Institute

2

ABSTRACT

Cancer vaccines are a type of therapy that uses a patient’s immune system (or components

thereof) to fight the patient’s tumor. Several types of cancer vaccines have been developed, including the

use of therapeutic antibody vaccines, dendritic cell vaccines, tumor-infiltrating lymphocyte vaccines,

chimeric antigen receptor vaccines, and immune checkpoint vaccines. While early vaccine experiments

often failed, recent studies have shown some spectacular successes. The goal of this IQP was to

investigate the field of cancer vaccines, assessing its problems, identifying future trends, and making

recommendations for moving the field forward. Our team performed a review of the current research

literature, and conducted interviews with scientists and physicians who design or use these vaccines. We

found the cancer vaccine field to be complex, but it provides a variety of approaches for potentially

treating a patient’s tumor. In some cases these approaches have already provided complete remissions for

relapsing cancers that would be impossible to treat using any other approach. While each type of therapy

can induce side-effects in a portion of the patients, we agree with our interviewees that the side-effects are

usually minor, transient, and treatable, and are far less severe than attempting to save the patient’s life

from a relapsing fatal cancer.

Page 3: CANCER VACCINES - Worcester Polytechnic Institute

3

TABLE OF CONTENTS

Title Page ……………………………….……………………………………..……. 01

Abstract …………………………………………………………………..…………. 02

Table of Contents ………………………………………………………………..… 03

Acknowledgements …………………………………………………………..…….. 04

Authorship ………………………………………………………….………….…… 06

Project Goals ………………………………………………………………………. 07

Background …………….……………………………………...….………….……. 09

Section-1: Introduction to Cancer ……………………...………………….. 09

Section-2: Introduction to Immunology …………………………………… 12

Section-3: Introduction to Cancer Vaccines ………………………………. 16

Literature Review …………………………………………………………………. 19

Section-1: Therapeutic Antibody Vaccines ……………………………….. 19

Section-2: Dendritic Cell (DC) Vaccines …………….……….………….. 48

Section-3: Tumor-Infiltrating Lymphocyte (TIL) Vaccines ……………… 60

Section-4: Chimeric Antigen Receptor (CAR) Vaccines …….……………. 67

Section-5: Immune Checkpoint Vaccines ………………………………… 79

Methods …………………………………………………………………………… 97

Results/Findings ……………………………..…………………………….……… 99

Conclusions and Recommendations ………..…………..…………………….…… 112

Appendix ……………………………………………………………………….…. 116

Sample Questions …………………….….…………………………….…. 116

Interview Preamble …………………………………………………….…. 117

Page 4: CANCER VACCINES - Worcester Polytechnic Institute

4

ACKNOWLEDGEMENTS

This IQP would not have been possible without the support of the individuals listed below. We

thank the following individuals for allowing us to interview them for this IQP project (alphabetical order

by last name):

Dr. Eduard Batlle. Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of

Science and Technology, Baldiri i Reixac 10, 08028 Barcelona, Spain.

Dr. Emily E. Bosco, PhD. Scientist-II, Oncology Research, MedImmune, LLC, Gaithersburg, Maryland

20878, USA.

Dr. Yoon Jin Cha. Department of Surgery, Gangnam Severance Hospital, Yonsei University College of

Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, Republic of Korea.

Dr. Chul Won Choi, MD. Department of Radiation Oncology, Dongnam Institute of Radiological &

Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan, 46033, Korea.

Dr. Eleonora DeMartin. Centre Hépatobiliaire, Hôpital Paul Brousse, Groupe Hospitalier Paris Sud,

DHU Hepatinov, RHU Ilite, 12 Avenue Paul Vaillant Couturier, 94800 Villejuif, France.

Dr. Andreas Engert. Professor for Internal Medicine, Hematology & Oncology, Department of Internal

Medicine, University Hospital of Cologne, Kerpener Str 62, 50937 Cologne, Germany. And Chairman of

the German Hodgkin Study Group.

Dr. Vancheswaran Gopalakrishnan. Department of Surgical Oncology, The University of Texas MD

Anderson Cancer Center, Houston, TX 77030, USA.

Dr. Steven C. Katz, MD, FACS. Associate Professor of Surgery, Director, Complex General Surgical

Oncology Fellowship, Director, Office of Therapeutic Development, Roger Williams Medical Center,

825 Chalkstone Avenue, Prior 4 Providence, Rhode Island 02908.

Dr. Linda M. Liau, MD. University of California Los Angeles (UCLA) David Geffen School of

Medicine & Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.

Dr. Christopher H. Lieu, MD. Director, GI Medical Oncology, and Deputy Associate Director for

Clinical Research, University of Colorado Anschutz Medical Campus, Division of Medical Oncology,

1665 Aurora Ct. Mail Stop F-703 | Aurora, Colorado 80045.

Dr. Michael Lim, MD. Professor of Neurosurgery, Oncology, Radiation Oncology, Otolaryngology, and

Institute of NanoBiotechnology, Director of the Brain Tumor Immunotherapy Program, Director of the

Metastatic Brain Tumor Center, Johns Hopkins University School of Medicine, 600 N. Wolfe Street,

Neurosurgery - Phipps 123, Baltimore, MD 21287.

Dr. Frederick L. Locke, MD. Department of Blood and Marrow Transplantation, Moffitt Cancer

Center, Tampa, FL 33612, USA.

Dr. Andrés Morales La Madrid, MD. Unidad de Neuro Oncología Pediátrica, Servicio de Oncología y

Hematología Pediátrica, Hospital St Joan de Déu, Passeig St Joan de Déu, 2, 08950 Esplugues de

Llobregat, Barcelona, Spain. Also: Laboratory of Developmental Cancer, Institut de Recerca Sant Joan

de Déu, Barcelona, Spain.

Page 5: CANCER VACCINES - Worcester Polytechnic Institute

5

Dr. Michael C. Milone. Center for Cellular Immunotherapies, Perelman School of Medicine at the

University of Pennsylvania, Philadelphia, Pennsylvania.

Dr. Michael A. Postow. Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College,

New York.

Dr. Chunjian Qi, MD, PhD. Professor and Director, Medical Research Center, The Affiliated

Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China; Oncology

Institute, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou,

213003, China.

Dr. Sergey E. Sedykh. Laboratory of Repair Enzymes, Siberian Branch of Russian Academy of

Sciences Institute of Chemical Biology and Fundamental Medicine, Novosibirsk State University,

Novosibirsk, Russia.

Dr. Erlin Sun. Department of Urology, Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical

University, Tianjin 300211, PR China.

Dr. Jakub Svoboda. Lymphoma Program, Abramson Cancer Center, University of Pennsylvania,

Philadelphia, PA.

Dr. John M. Timmerman. Division of Hematology & Oncology, Department of Medicine, Center for

Health Sciences, Room 42-121, University of California at Los Angeles, 10833 LeConte Avenue, Los

Angeles, CA 90095-1678.

Dr. Marek Trněný, MD, CSc. Professor and Chairman, 1st Dept Medicine, 1st Fac Medicine, Charles

University, General Hospital, U nemocnice 2, CZ 128 08 Praha, Czech Republic.

Dr. L.I. Zon. Harvard Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute,

Harvard University, 7 Divinity Ave (Fairchild Building, Room G53), Cambridge, MA 02138.

Dr. Emese Zsiros. Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center,

Buffalo, NY, USA; and Center for Immunotherapy, Roswell Park Comprehensive Cancer Center,

Buffalo, NY, USA.

Anonymous. Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital,

No.123, Da-Pei Road, Niaosung Dist, Kaohsiung City, Taiwan, R.O.C.

Anonymous. Departments of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel

School of Medicine at Dartmouth, 621 Rubin Building - HB7936; 1 Medical Center Drive, Lebanon, NH,

03756, USA.

Anonymous. Thyroid Cancer Group, Ingham Institute for Applied Medical Research, Liverpool, NSW,

Australia; School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.

In addition to the interviewees listed above, we would also like to thank Dr. David Adams for

serving as project advisor for this IQP. We found Dr. Adams to be invaluable from the very onset of this

project, through concept initiation and managing the team, to the final conclusions. We thank him for his

enthusiasm, dedication, and continued guidance.

Page 6: CANCER VACCINES - Worcester Polytechnic Institute

6

AUTHORSHIP

Author Topics Covered

Johan Girgenrath Therapeutic Antibody Vaccines

Leila Camplese Dendritic Cell Vaccines

Michaela Hunter Tumor-Infiltrating Lymphocyte Vaccines

Talal Hamza Chimeric Antigen Receptor Vaccines

Cole Royer Checkpoint Inhibitor Vaccines

Page 7: CANCER VACCINES - Worcester Polytechnic Institute

7

PROJECT GOALS

Mission Statement and Objectives

Cancer is a collection of related diseases, all sharing the property of uncontrolled cell division. It

can initiate almost anywhere in the human body, and has over one-hundred different variations. At the

cellular level, cancer is a disease caused by DNA mutations and uncontrolled division of abnormal cells

in the body. As a result of these cellular changes, cancer can be difficult to treat. With our increasing

aging population, the number of people in the U.S. dying of cancer has risen over the years. But with

more accurate and sensitive detection techniques, better drugs for blocking cell growth (using a variety of

mechanisms), and more precise surgical tools for removing tumors, the death rates have actually declined

since 1990. Tumor treatments vary widely, depending on the location and type. Treatment options

include chemotherapy (the use of chemical drugs to block DNA replication or other key cancer processes,

including taxanes, anthracyclines, platins), radiation (to kill rapidly dividing cells), surgery (to remove the

tumor), targeted cancer therapy (drugs that interfere with specific molecules a tumor needs to grow), and

biologic therapy (i.e. cancer vaccines, the subject of this IQP).

But despite the advances mentioned above, and the large amount of money provided by the “War

on Cancer” (initiated by the National Cancer Act of 1971), cancer still remains the second leading cause

of death in the U.S., following cardiovascular disease, so better therapies are needed. The subject of this

IQP is a new form of targeted cancer therapy termed “cancer vaccines”. This type of therapy uses the

patient’s own immune system, or components of the immune system (such as antibodies, T-cells,

dendritic cells, etc.), to fight the patient’s tumor. Several types of cancer vaccines have been developed,

and each will be investigated in this IQP with respect to effectiveness and problems:

1) therapeutic antibodies against proteins present on the surface of tumor cells

(mono- specific antibodies, bi-specific antibodies, and antibody-drug conjugates),

2) dendritic cell vaccines,

3) tumor-infiltrating lymphocyte vaccines

4) chimeric antigen receptor vaccines

5) immune checkpoint vaccines

As is the case for all experimental therapies, cancer vaccines are approved for use in cancer

patients only if their tumors do not respond to traditional therapies such as chemotherapy or radiation

treatment. So, the patients assigned to cancer vaccine clinical trials are those with very poor prognosis.

Under these drastic conditions, any successes are worth pursuing. While the early cancer vaccines did not

work well, the past few years have shown some spectacular successes, including complete cancer

remissions for highly refractive tumors. The topic of cancer vaccines has become one of the hottest topics

in all of cancer research. Currently, researchers are racing to expand the use of immuno-therapies to

benefit different types of cancer patients. Hundreds of clinical trials are now underway to see whether

improved responses can be achieved by using combination therapies, each working with a different

mechanism. Advances in rapid and affordable DNA sequencing technologies have allowed the

identification of a patient’s own tumor neo-antigens as targets, creating a form of personalized medicine.

In addition, scientists have determined that the patient’s gut microbiome (type and number of microbes

present in the patient’s GI tract) helps determine whether the patient will respond to therapy.

But these new vaccines still come with problems. They usually cause deleterious side-effects,

they can be very expensive (especially for the personalized vaccines), and they do not work well in all

patients. The overall goal of this IQP project is to document and evaluate this new technology, to

determine which types of cancer vaccines work best, to document their problems, and help prioritize

future directions.

Page 8: CANCER VACCINES - Worcester Polytechnic Institute

8

The specific objectives are to:

1 Develop a comprehensive assessment of the scientific experiments that led to the development

and use of cancer vaccines.

2 Characterize what key scientific stakeholders believe are the strengths, weaknesses, reliability,

usefulness, and cost of this new technology, and any other concerns.

3 Evaluate all of the obtained evidence and prioritize the remaining problems.

4 Recommend potential solutions for remaining problems, and prioritize future experiments.

Notes on Project Risks

1. Potential Risk to the Human Subjects: The risks to the interviewees, if any, should be very

minor. For this type of project, the information requested and disclosed in the interviews will be

technical, or the interviewees own personal opinions. This type of information should not be

harmful if disclosed. This project has no need to request or disclose proprietary information, such

as technical secrets, as this is not needed to accomplish the goals of the project.

2. Justification of Risk: Interviews with human subjects (biomedical researchers) are required to

obtain information for this project beyond a standard review of the existing literature, although

the risk is minor due to the type of information disclosed.

3. Risk Reduction: For this project, risk to the interviewees will be reduced by informing the

subjects that the interview is voluntary (so they can withdraw their response if they feel it might

be risky to divulge the information asked for), they may end the interview at any time, and they

can ignore any question they wish. If we plan to quote an individual in our report, we will first

obtain the interviewee’s permission. Any request for confidentiality will be honored by making

an anonymous quote, or by not citing the information.

Page 9: CANCER VACCINES - Worcester Polytechnic Institute

9

BACKGROUND

Section-1: Introduction to Cancer

Cancer Description and Causes

Cancer is a collection of related diseases, all sharing the property of uncontrolled cell division. It

can initiate almost anywhere in the human body, and has over one-hundred different variations. The extra

cells form growths called tumors. Some cancers form solid tumors, while others, such as leukemia, form

diffuse tumors. Malignant tumors spread into (invade) nearby tissues, or can break off and travel to

distant places in the body through the blood or the lymph system to form new tumors. Benign tumors do

not invade nearby tissues. From the prostate gland, to the thyroid glands, the lungs, or the kidney, these

malignancies symptomatically grow and strongly affect health.

At the cellular level, cancer is a disease caused by DNA mutations and uncontrolled division of

abnormal cells in the body. It has several hallmarks, including: genomic instability, deregulated cell

signaling causing cell division and growth, sustained cell proliferation, resistance to cell death, and

evasion from the patient’s immune system (Hanahan and Weinberg, 2011). As a result of these cellular

changes, cancer can be difficult to treat.

Cancer Prevalence and Survival Rates

According to the Centers for Disease Control, in 2015 (their most recent data) cancer was the

second leading cause of death in the United States, with 595,930 (22.7%) deaths, compared to 633,842

(24.1%) for the number-1 killer heart disease (CDC, 2018). The number of people in the US dying of

cancer has risen over the years with our increasing aging population, but with improved cancer treatments

the death rates have actually declined since 1990 (CBS, 2018). The gap between the number-1 killer and

number-2 killer is decreasing, likely due to the improving survival rates for heart disease patients (CBS,

2018).

Cancer Survival Rates

Cancer survival rates vary widely, from 8%-18% for difficult to treat cancers, such as pancreatic,

lung, or liver cancers, to greater than 65% for easier to treat cancers, such as cancers of the colon, breast,

kidney, and prostate (that grow slowly and are easier to treat if detected early) (Howlader et al.,

2017). Cancer survival rates have improved significantly in the past several decades due to more accurate

and sensitive detection techniques, better drugs for blocking cell growth (using a variety of mechanisms),

and more precise surgical tools to remove tumors.

Page 10: CANCER VACCINES - Worcester Polytechnic Institute

10

Cancer Treatments

Tumor treatments vary widely, depending on the location and type. Benign tumors in a safe area

of the body that do not cause organ disruption are sometimes left alone and watched carefully. If a tumor

begins uncontrolled growth, treatment options include chemotherapy (the use of chemical drugs to block

DNA replication or other key cancer processes, including taxanes, anthracyclines, platins), radiation (to

kill rapidly dividing cells), surgery (to remove the tumor), targeted cancer therapy (drugs that interfere

with specific molecules a tumor needs to grow), and biologic therapy (i.e. cancer vaccines, the subject of

this IQP).

Strong Need for New Cancer Drugs

Despite the advances mentioned above, and the large amount of money provided by the “War on

Cancer” (initiated by the National Cancer Act of 1971), cancer still remains the second leading cause of

death (following cardiovascular disease), so better therapies are needed.

Cited References on Cancer

American Cancer Society (2018) Cancer A to Z. https://www.cancer.org/cancer.html

American Lung Association (2018) Lung Cancer Fact Sheet. http://www.lung.org/lung-health-and-diseases/lung-disease-lookup/lung-cancer/resource-library/lung-

cancer-fact-sheet.html Bajetta E, Del Vecchio M, Bernard-Marty C, Vitali M, Buzzoni R, Rixe O, Nova P, Aglione S, Taillibert

S, Khayat D (2002) Metastatic melanoma: chemotherapy. Seminars in Oncology, 29 (5): 427–445.

Cancer.org (2018) Prostate Cancer Key Statistics. https://www.cancer.org/cancer/prostate-cancer/about/key-statistics.html

Cancer.org (2018) Breast Cancer. https://www.cancer.org/cancer/breast-cancer.html CBS News (2018) The Top Leading Causes of Death in the US. https://www.cbsnews.com/news/the-

leading-causes-of-death-in-the-us/ CDC.org (2018) Leading Causes of Death. Data are for 2015. https://www.cdc.gov/nchs/fastats/leading-causes-of-death.htm

Damber JE, Aus G (2008) Prostate Cancer. Lancet, 2008 May 17; 371(9625): 1710-1721. Dong JT (2006) Prevalent mutations in prostate cancer. Journal of Cellular Biochemistry, 2006 Feb 15;

97(3): 433-447. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell, 2011 Mar 4; 144(5):

646-674.

Healthline.com (2018) What Are the 12 Leading Causes of Death in the United States?

https://www.healthline.com/health/leading-causes-of-death#1

Page 11: CANCER VACCINES - Worcester Polytechnic Institute

11

Leukemia Research Foundation (2018) Leukemia. http://www.allbloodcancers.org/statistics National Cancer Institute (2018) Leukemia. https://www.cancer.gov/types/leukemia Noone AM, Howlader N, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis

DR, Chen HS, Feuer EJ, Cronin KA (editors) (2018) SEER Cancer Statistics Review, 1975-2015,

National Cancer Institute. Bethesda, MD. Updated 4-16-2018. https://seer.cancer.gov/csr/1975_2015/

Page 12: CANCER VACCINES - Worcester Polytechnic Institute

12

Background Section-2:

Introduction to Immunology

This IQP focuses on the immune system, so a brief introduction to this topic will aid our

understanding of how cancer vaccines work. The immune system is composed of a network of cells,

tissues, and organs whose main purpose is to protect the body from disease and infection. Cells in the

immune system recognize problems in the body, communicate with other cells, and react to perform

beneficial functions. The immune system is divided into two major sub-divisions: innate immunity and

adaptive immunity.

Main Divisions of the Immune System

Innate immunity is the portion of the immune system that is ready for immediate response when

an infection is first detected. This system includes physical and chemical surface barriers (such as the

skin, sweat, tears, saliva, respiratory tract mucous, stomach acid, and urine) which serve as an initial

barrier to infection (Science Learning Hub, 2010). This system also includes the use of defensive cells,

defensive proteins, inflammation, and fever. The cells involved in innate immunity include: natural killer

cells (NKs), mast cells, eosinophils, basophils, and phagocytic cells [macrophages, neutrophils, and

dendritic cells (DCs)]. These cells recognize molecular patterns present on the surface of bacteria and

fungi, and act to engulf the pathogens (or aid other cells that engulf and kill them) (Vesely et al., 2011).

DCs are also part of the body’s adaptive immune system (see below). If a pathogen is able to survive the body’s innate defenses, the body will eventually react with a

more advanced response to specifically target the pathogen. This adaptive immune system is also known

as the antigen-specific immune response (Spurrell and Lockley, 2014). Antigens are short domains of

amino acids or sugars that are viewed as foreign by the immune system. The adaptive cells of this system

include antigen-presenting cells (APCs), B-lymphocytes (B-cells), and T-lymphocytes (T-cells). APCs

make contact with a pathogen, internalize it, and process selected antigens for presentation on the cell

surface. These presented antigens bind to immature pre-B-cells and pre-T-cells, which then begin to

differentiate and commit to the antigen. Once these cells have matured, they are specific to the antigen

that induced them: B-cells manufacture and secrete antibodies against the antigen into the blood, and

cytotoxic T-cells identify infected cells and kill them. DCs, B-cells, and T-cells are all part of the cancer

vaccine topic (National Cancer Institute, 2018). Some cancer vaccines use DCs isolated from a patient to

prime them against a tumor-specific antigen, then perfuse the DCs back into the patient to induce B-cells

and T-cells to eliminate the tumor. Other cancer vaccines isolate, prime, and perfuse T-cells back into the

patient.

Antigen Presenting Cells (APCs)

As their name implies, APCs are a specialized type of white blood cell that present foreign

antigens on their surface. These presented antigens are recognized by other components of the immune

system, such as B-cells and T-cells, to help them commit to that specific antigen (Wellness, 2015). In this

process, a foreign invader is detected by an APC and engulfed. Proteases inside the APC degrade foreign

antigens on the invader surface into smaller peptides which are then transported to the APC surface where

they combine with either an MHC type-I molecule (professional presentation) or a type-II molecule

(non-professional presentation). This antigen-MHC complex is then recognized by B-cells and T-cells

to help them commit to that particular antigen (Kimball’s Biology Pages, 2013). Professional APCs

Page 13: CANCER VACCINES - Worcester Polytechnic Institute

13

include dendritic cells, macrophages, or B-cells. These cells present their antigens using MHC-I, and are

the only type to activate helper T-cells. Non-professional APCs include fibroblast cells, thymus epithelial

cells, thyroid epithelial cells, glial cells, pancreatic beta cells, and vascular endothelial cells. These cells

present antigens using MHC-II in a weaker type interaction (Garland Science, 2001).

Antibodies

One type of cancer vaccine involves injecting the patient with antibodies directed against the

patient’s tumor cells. Antibodies are proteins secreted by mature B-cells into the bloodstream that

interact with foreign invaders to bring them to the attention of the immune system for

elimination. Antibodies have a “Y” structure (Figure-1) comprised of two long chains and two short

chains. The antibody constant regions (blue in the diagram) dictate which type of immune cell the

antibody engages, while the variable domains (red in the diagram) interact with the foreign antigen.

Figure-1: Diagram of a Typical Antibody Structure. Antibodies have a

general structure that is similar to a “Y”. It contains variable domains (red)

that specifically bind to the foreign antigen, and constant domains (blue) that

dictate which of the body’s immune cells are engaged by the antibody (Murphy

and Weaver, 2016).

The antibody variable region that binds the antigen is unique to each antibody clone (the group of

antibody molecules secreted from a mature plasma cell and all its derivatives). It has been estimated that

the human immune system can produce over 5 x 1013 different types of antibodies (Murphy and Weaver,

2016). The constant regions of the antibody molecules dictate which class the antibody belongs to. There

are five main classes of antibodies: IgA, IgD, IgE, IgG, and IgM. The constant regions remain fairly

conserved within within each class (Vidarsson et al., 2014). The constant region allows the antibody

molecule to interact with effector molecules and phagocytic cells that internalize the antibody-antigen

complex.

Once an antibody binds its antigen, based on its type of constant region, it signals for a specific

effector function to help destroy the pathogen. There are three main paths for destruction: 1)

neutralization (an antibody blocks a binding site keeping the pathogen from entering a cell, 2)

opsonization (the antibody-antigen complex is taken inside a macrophage cell for destruction), or 3)

complement system activation by the constant region (this system signals plasma proteins to bind to and

puncture the pathogen’s membrane, leading to cell lysis, and coats the pathogen’s membrane to attract

phagocytic cells.

Page 14: CANCER VACCINES - Worcester Polytechnic Institute

14

B-Lymphocytes (B-Cells) B-lymphocytes (B-cells) are produced in the bone marrow, the tissue from which they derive

their name. Pre-B-cells recognize a foreign antigen presented by an antigen-presenting cell

(Immunobiology, 2001). This interaction initiates a maturation process that commits the B-cell to

producing antibodies against the antigen. Cytokine hormones and helper T-cells aid the maturation

process, which results in a plasma cell that secretes antibodies. The committed plasma or B-cells are then

clonally expanded to increase their numbers.

Dendritic Cells (DCs)

Dendritic cells (DCs) derive their name from their branched appearance at specific stages of their

development. They are potent “professional” antigen-presenting cells whose main function is to recognize

foreign antigens (usually small epitope domains of proteins) on the surface of invading pathogens (and

sometimes cancer cells), process the antigen within the cell, and then present it on its surface to other cells

of the immune system, such as T-cells and B-cells, so they can help eliminate the tumor. The B-cells and

T-cells interact with the presented antigen to commit to it. Half of the 2011 Nobel Prize in Physiology or

Medicine went to Ralph M. Steinman for “his discovery of the dendritic cell and its role in adaptive

immunity" (The Nobel Prize, 2011). Because of their ability to present antigens to the immune system,

DCs are used in some types of cancer vaccines to induce a patient’s immune response against an antigen

on the surface of a patient’s tumor cell.

T-Cells

T-lymphocytes (T-cells) are a type of nucleated white blood cell that functions in adaptive

cellular immunity. T-cells are distinguished from other lymphocytes, such as B-cells and natural killer

cells (NK cells), by the presence of a T-cell receptor (TCR) on their cell surface which recognizes a

presented antigen and commits the cell against that antigen (Immunobiology, 2001). They are called T-

cells because they mature in the thymus (although some T-cells also mature in the tonsils). Several types

of T-cells exist, each with a different function: helper (CD4+), cytotoxic (CD8+), memory, suppressor,

mucosal, and gamma delta T-cells.

With respect to the topic of cancer vaccines, tumor infiltrating lymphocytes (TILs) are a type

of T-cell found in tumors that help kill it. High levels of TILs in tumors are often associated with a better

clinical outcome for the patient. TILs isolated from tumors usually include both CD4+ (helper T-cells) and

CD8+ (cytotoxic killer T-cells, CTLs). TILs circulate through the bloodstream, recognize the tumor and

infiltrate it. The CD4+ helper T-cells secrete cytokines to boost the immune system. CTLs directly lyse

the tumor cell.

Cited References on Immunology

Cancer Research UK (2018) The immune system and cancer. http://www.cancerresearchuk.org/about-cancer/what-is-cancer/body-systems-and-cancer/the-immune-

system-and-cancer

Immunobiology (2001) The Immune System in Health and Disease. 5th Edition. Chapter-3: Antigen

Recognition by B-Cell and T-Cell Receptors. National Institutes of Health.

Page 15: CANCER VACCINES - Worcester Polytechnic Institute

15

https://www.ncbi.nlm.nih.gov/books/NBK10770/ Murphy K, Weaver C (2016) Janeway’s Immunobiology. 9th Ed. New York NY.

National Cancer Institute (2018) Immunotherapy: Using the Immune System to Treat Cancer. Site was

updated on 2-14-18. https://www.cancer.gov/research/key-initiatives/immunotherapy. Schultz KT, Grieder F (1987) Structure and function of the immune system. Toxicologic Pathology, 1987;

15(3): 262-264.

Science Learning Hub (2010) The Body’s First Line of Defense. Updated 11-2-2010. https://www.sciencelearn.org.nz/resources/177-the-body-s-first-line-of-defence.

Spurrell EL, Lockley M (2014) Adaptive immunity in cancer immunology and therapeutics. E-Cancer Medical Science, 8: 441. DOI: 10.3332/ecancer.2014.441. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4096025/

The Nobel Prize in Physiology or Medicine (2011) https://www.nobelprize.org/nobel_prizes/medicine/laureates/2011/#

Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ (2011) Natural innate and adaptive immunity to

cancer. Annual Review of Immunology, 2011; 29: 235-271. doi: 10.1146/annurev-immunol-031210-101324.

Vidarsson G, Dekkers G, Rispens T (2014) IgG Subclasses and Allotypes: From Structure to Effector

Functions. Frontiers in Immunology, 2014; 5: 520.

Wellness (2018) Antigen-Presenting Cells. https://www.wellness.com/reference/allergies/antigen-

presenting-cells.

Page 16: CANCER VACCINES - Worcester Polytechnic Institute

16

Background Section-3:

Introduction to Cancer Vaccines

The subject of this IQP is a form of targeted cancer therapy, cancer vaccines. This type of

therapy uses the patient’s own immune system, or components of the immune system (such as antibodies,

T-cells, dendritic cells, etc.), to fight the patient’s tumor. Several types of cancer vaccines have been

developed, and each will be investigated in this IQP: 1) injecting cancer neo-antigens (peptides specific to the cancer cell surface) to help the

immune system make antibodies and T-cells against the tumor, 2) injecting therapeutic antibodies against proteins on the surface of tumor cells (mono-

specific antibodies, bi-specific antibodies, and antibody-drug conjugates), 3) injecting dendritic cell vaccines, 4) injecting T-cell vaccines (TILs and CARs), and 5) injecting immune system modulators (such as immune checkpoint inhibitors).

As is the case for all experimental therapies, cancer vaccines are approved for use in cancer

patients only if their tumors do not respond to traditional therapies such as chemotherapy or radiation

treatment. So, patients assigned to cancer vaccine trials tend to be those with very poor prognosis.

Although the early cancer vaccines did not work well, the past few years have shown some spectacular

successes, including some complete cancer remissions for highly refractive tumors. Thus, the topic of

cancer vaccines has become one of the hottest topics in all of cancer research.

The past few years have seen unprecedented clinical successes, rapid drug developments, and

“first-in-kind” treatment approvals from the FDA. In 2016, the American Society of Clinical Oncology

(ASCO) announced “immunotherapy” as the year's top cancer advance, and in 2017 that society named

immunotherapy as its “advance of the year” (Madden, 2018). The society emphasized the rapid pace of

research in this field, emphasizing that “these agents have extended the lives of many patients with late-

stage cancers for which there have been few treatment options” (Madden, 2018). In 2017, approximately

2,000 immuno-therapeutic agents were under development (Schmidt, 2017).

Researchers are racing to expand the use of immuno-therapies to benefit more types of cancer

patients. Hundreds of clinical trials are now underway to see whether improved responses can be achieved

by using a combination of two immunotherapies, each working with a different mechanism. The

number of clinical trials is increasing at an exponential pace, as evidenced by the number of combination

trials with checkpoint inhibitors and another treatment (Figure-2).

Figure-2: Increase in Cancer

Vaccine Clinical Trials. Shown is an

example of the exponential increase in

clinical trials combining a checkpoint

inhibitor vaccine with another

treatment. Diagram is from Schmidt,

2017.

Page 17: CANCER VACCINES - Worcester Polytechnic Institute

17

Advances in rapid and affordable DNA sequencing technologies have allowed the identification

of a patient’s own tumor neo-antigens as targets, creating a form of personalized medicine. In addition,

scientists have found that the patient’s gut microbiome (type and number of microbes present in the

patient’s GI tract) helps determine whether the patient will respond to therapy.

The breadth of cancers treatable with cancer vaccine combinations has increased in recent years

(Figure-3). Lung cancer, melanoma, breast cancer, lymphoma, kidney cancer, and head and neck cancers

are among the most researched cancers treated with immuno-vaccines.

Figure-3: Main Types of Cancer Treated with Vaccines. Shown are the most

common cancers treated with cancer vaccine combination trials. Diagram is from

Schmidt, 2017.

Problems with Cancer Vaccines While recent years have shown some spectacular successes with cancer vaccines, much still needs

to be done, as these new cancer vaccines come with problems:

1) It remains unclear why only a subset of patients respond to a particular therapy. Does the tumor stop

making the antigen targeted by the therapy, allowing re-growth? Are components of the patient’s immune

system blocking the success of the therapy?

2) Why are the immuno-therapies so expensive? A recent study indicates that the average checkpoint

vaccine in the US costs $150,000, and the average CAR vaccine about $475,000 (Couzin-Frankel,

2018). Who should pay the price for such expensive medicines? 3) Why do most cancer vaccines induce side-effects? Are the side-effects transient and manageable? The overall goal of this IQP is to document and evaluate the technology of cancer vaccines, to document

technique problems and help prioritize future directions.

Cited References for Introduction to Cancer Vaccines

Couzin-Frankel J (2018) Sticker shock. Science, 2018 Mar 23; 359(6382): 1348-1349. doi: 10.1126/science.359.6382.1348.

Page 18: CANCER VACCINES - Worcester Polytechnic Institute

18

Madden DL (2018) From a Patient Advocate's Perspective: Does Cancer Immunotherapy Represent a

Paradigm Shift? Current Oncology Reports, 2018 Feb 7; 20(1): 8. doi: 10.1007/s11912-018-0662-5.

Schmidt C (2017) Combinations on Trial: By Giving Immunotherapies in Tandem, Researchers Hope to

Broaden the Benefits. Nature, 552: S67–S69. The data in the article was updated on January 10, 2018

in Nature, 553: 155, to correct the number of immunotherapeutic agents in development from

2,400 down to 2,000. doi: 10.1038/d41586-018-00415-9.

Page 19: CANCER VACCINES - Worcester Polytechnic Institute

19

LITERATURE REVIEW

Section-1: Therapeutic Antibody Vaccines Johan Girgenrath

Cancer Antigens and Neo-Antigens as Antibody Targets

The success of all cancer vaccines depends on the existence of antigens (proteins or sugars, or

portions thereof) on the surface of the cancer cells that can be specifically targeted by the

vaccine. Tumors are caused by mutations in DNA. Some of these mutations alter the expression levels or

types of antigens on the surface of the tumor (Parmiani et al., 2007). New antigens presented on the

tumor surface that are lacking in normal cells are termed neo-antigens (Schumacher and Schreiber,

2015). Neo-antigens can vary from tumor to tumor, and from patient to patient, so they are the subject of

much research in the personalized medicine field.

Tumor cells in the body are poor antigen-presenting cells. Tumor cells are derived from normal

cells by DNA mutation, so the vast majority of the tumor cell DNA is identical to the patient’s normal

cells. Thus, the tumor surface antigens look mostly like “self” to the immune system, and are ignored,

allowing the tumor to grow. Only a small portion of the cancer DNA mutations create neo-antigens

unique to the patient’s tumor, and these provide excellent candidates for cancer vaccine designs. One of

the goals of cancer vaccine research is to develop rapid affordable methods for determining the exact neo-

antigens present in a specific patient’s tumor, and designing a personal vaccine for that patient.

Much research in the cancer vaccine field has focused on identifying specific antigens for

targeting. These antigens should not be found in large quantities in normal cells, to prevent their damage

by the vaccine. Examples include proteins CD19, CD20, or CD22 on the surface of B-cells, which are

targeted by cancer vaccines against B-cell tumors (such as leukemia), overactive B-cells (autoimmune

disorders, transplant rejection), or for killing dysfunctional B-cells. Another well-known example is the

protein Her-2, which is over-expressed on some types of breast cancer cells, and is targeted by the

monovalent cancer vaccine antibody Herceptin (also known as Trastuzumab or Herclon).

Mono-Specific Antibody Vaccines

One type of cancer vaccine consists of injecting the patient with antibodies against the tumor

cell. As mentioned in the Immunology Introduction section, antibodies are proteins secreted by mature

B-cells (plasma cells) into the bloodstream that interact with foreign invaders to bring them to the

attention of the immune system for elimination. Injecting antibodies into a patient is termed passive

immunity. It does not activate the patient’s own immune system to create the antibodies, but instead the

antibodies are produced by a bio-engineering process. The antibodies bind to antigen to create antigen-

antibody complexes, which are then recognized and cleared from body by other cells of the immune

system, such as macrophages or T-cells. The antibodies used in a cancer vaccine can be mono-specific,

bi-specific, or antibody-drug conjugates.

Mono-specific (or monovalent) antibodies recognize only one type of antigen. Most natural

antibodies produced in the human body against an infection are of this type. The variable domains on

both arms of the “Y” shaped antibody recognize the same antigen. For example, the first antibody

Page 20: CANCER VACCINES - Worcester Polytechnic Institute

20

approved as a cancer vaccine in the U.S. was Rituximab, an antibody against surface protein CD20. This

protein is found on the surface of B-cells, so the antibody is used to treat patients with high B-cell

numbers (leukemia and lymphoma), patients with overactive B-cells (autoimmune disorders, transplant

rejection), or patients with dysfunctional B-cells. Rituximab was initially approved by the FDA in 1997

to treat non-Hodgkin (Maloney et al., 1997). Several studies have used it in clinical trials with various

success. Another example is Inotuzumab, a mono-specific antibody against CD22, used to treat patients

with refractory acute lymphoblastic leukemia (ALL). Perhaps the best known antibody in this category is

Herceptin (also known as Trastuzumab or Herclon), the second antibody approved by the FDA for cancer

treatment in the U.S. Herceptin binds to the HER2/neu receptor, which is over-expressed on some types

of cancer cells (Bange et al., 2001). Herceptin was approved by the FDA in September 1998 for treating

HER2-positive breast cancers, and is now used to treat colorectal and pancreatic cancers (Perez et al.,

2002).

Example of Mono-Specific Antibody Clinical Trials

With respect to clinical trials using mono-specific antibodies, although the treatments in this

category sometimes prolonged a patient’s life, a review of the literature showed that full cancer

remissions have not been that common. Table-I below shows examples of clinical trials done with

mono-specific antibodies.

Table-I: Example Clinical Trials With Mono-Specific Antibody Vaccines

Target

Antigen Cancer Notes Side-Effects Reference

CD20

Low-grade

non-

Hodgkin’s

lymphoma

Phase-III trial on 116 patients, 48%

achieved a measurable tumor response, 6%

achieved complete tumor responses, 76%

achieved ≥20% reduction in tumor volume.

Berinstein et

al., 1998

Indolent B-

cell

lymphoma

Combination trial of Rituximab with

chemotherapy, 40 patient group, 55%

complete remission and 40% partial

remission.

Czuczman,

1999

Stage II-IV

low-grade

non-

Hodgkin’s

lymphoma

39 patients, 54% of the patients showed

objective responses, at 1 year 77% showed

progression-free survival.

The treatment was

well tolerated. Hainsworth,

2000

Aggressive

non-

Hodgkin’s

lymphoma

(NHL)

Rituxan plus chemotherapy combination, 33

patients, 61% of the patients experienced a

“complete response”. 29 of 31 responding

patients remained in remission during a 26

month follow-up period.

The most frequent

adverse events

attributed to the

Rituxan antibody

were fever and chills.

Vose et al.,

2001

Non-Hodgkin

lymphoma

(NHL)

Rituximab, 50 patients, the response rate

after 50 days was 73%, with 10 patients

(20%) in complete remission, 3 patients in

complete remission/unconfirmed, and 23

patients in partial remission.

Colombat et

al., 2001

Page 21: CANCER VACCINES - Worcester Polytechnic Institute

21

Indolent non-

Hodgkin’s

lymphoma

62 patients, Following a second Rituximab

treatment, the major response rate increased

from 47% to 65%, and the complete

response rate increased from 7% to 27%.

There was no

observable toxicity

with repeat courses of

rituximab.

Hainsworth,

2002

HER-2

Metastatic

breast cancer 222 women, blinded, independent response

evaluation committee identified 8 complete,

and 26 partial responses, providing an

overall response rate of 15%.

The most common

adverse events were

infusion-associated

fever and/or chills

(40% of the patients).

Cobleigh et

al., 1999

Metastatic

breast cancer 234 patients were randomly assigned to

receive standard chemotherapy, and 234

patients were assigned to receive standard

chemotherapy plus trastuzumab. The results

indicated that the addition of trastuzumab

antibody to the chemotherapy regime

provided a longer time to disease

progression (median 7.4 vs 4.6 months;

p<0.001) and a lower rate of death at 1 year

(22% vs 33%, p=0.008).

The most important

adverse event

observed was cardiac

dysfunction (27% in

chemo group versus

13% in the

combination group).

The symptoms were

manageable.

Slamon et

al., 2001

Advanced

breast cancer An international, multicenter, randomized

trial, 1,694 received trastuzumab and 1,693

were controls. At one year, recurring breast

cancer or death was observed in 127 patients

in the trastuzumab group versus 220 in the

control group.

Severe cardiotoxicity

developed in 0.5% of

the women treated

with trastuzumab.

Piccart-

Gebhart et

al., 2005

Breast cancer

patients

27 breast cancer patients were treated with

trastuzumab and chemotherapy. Anti-HER-

2/neu antibodies were detectable in 29% of

the patients before treatment, and in 56% of

the patients during treatment. Of the

twenty-two individuals treated for metastatic

disease, those showing improved clinical

responses had higher levels of HER-2

antibodies.

Taylor et al.,

2007

CD22

Acute

lymphocytic

leukemia

(ALL)

Phase-II clinical trial. The antibody was

conjugated to the toxin calecheamicin to kill

the CD22+ cells. Of the 49 treated patients:

9 (18%) had a complete response, 19 (39%)

had resistant disease, and only 2 (4%) died.

The most frequent

side effects were

fever (20 patients),

hypotension (12

patients), and liver

toxicity (12 patients).

Kantarjian et

al., 2012

Page 22: CANCER VACCINES - Worcester Polytechnic Institute

22

Acute

lymphocytic

leukemia

(ALL)

Follow-up study, Inotuzumab (a CD22 mAb

bound to toxin calicheamicin), 90 patients,

49 received a single-dose 3-4 weeks, while

41 patients received inotuzumab weekly

every three to four weeks. The overall

response rate was 58%, 19% achieved a

complete response, 30% had a complete

response with no platelet recover (CRp), and

9% had a bone marrow complete recovery.

The response rates were similar between the

two groups.

Some of the adverse

side effects observed

were reversible

bilibrubin elevation,

fever, and

hypotension.

Kantarjian et

al., 2013

EGFR

Non-small-

cell lung

cancer

(NSCLC)

Phase III clinical trial. Treatment with mAb

Cetuximab (binds EGFR) + chemotherapy

(n = 557) vs treatment with chemotherapy

(n = 568). Patients treated with both

cetuximab and chemotherapy survived

several months longer than patients treated

solely with chemotherapy.

10% of patients that

were administered

cetuximab observed

an acne-like rash.

Pirker et al.,

2009

Problems with Monovalent Vaccines

Because tumor cells are mutated patient cells, there are still many similarities between normal

patient cells and tumor cells. As a result, monovalent antibodies that are designed against antigens present

on a tumor cell may also recognize these same antigens on healthy cells, disrupting normal function and

causing off-target effects. In addition, some tumors downregulate expression of the target antigen, or

don’t express it at all. For example, in B-cell cancers, CD22 is present on about 60-90% of B-cell

malignancies, but not in 10-40% of leukemic patients (Hoelzer, 2013). In spite of this, some success has

been achieved with CD19 antibodies for B-cell tumors and leukemia (Naddafi et al., 2015), likely because

CD19 is restricted to the B-cell lineage, and lost B-cells can be replaced in the patient post-cancer

treatment. To be effective, monovalent antibody vaccines need a healthy patient’s immune system to help

clear the cancer cells tagged by the vaccine; binding of the antibody by itself does not kill the cancer cell.

So, monovalent vaccines might not work well in immuno-compromised patients.

References for Monovalent Vaccines

Arlen PM, Gulley JL, Parker C, Skarupa L, Pazdur M, Panicali D, Beetham P, Tsang KY, Grosenbach

DW, Feldman J, Steinberg SM, Jones E, Chen C, Marte J, Schlom J, Dahut W (2006) A randomized

phase II study of concurrent docetaxel plus vaccine versus vaccine alone in metastatic androgen-

independent prostate cancer. Clinical Cancer Research, 2006 Feb 15; 12(4): 1260-1269. Bange J, Zwick E, Ullrich A (2001) Molecular targets for breast cancer therapy and prevention. Nature

Medicine, 2001 May; 7(5): 548-552. Berinstein NL, Grillo-López AJ, White CA, Bence-Bruckler I, Maloney D, Czuczman M, Green D,

Rosenberg J, McLaughlin P, Shen D (1998) Association of serum Rituximab (IDEC-C2B8) concentration

and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkin's lymphoma.

Annals of Oncology, 1998 Sep; 9(9): 995-1001.

Page 23: CANCER VACCINES - Worcester Polytechnic Institute

23

Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, Aaronson SA (1991)

Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science,

1991 Feb 15; 251(4995): 802-804. Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, Fehrenbacher L, Wolter JM, Paton V, Shak S,

Lieberman G, Slamon DJ (1999) Multinational study of the efficacy and safety of humanized anti-HER2

monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has

progressed after chemotherapy for metastatic disease. Journal of Clinical Oncology, 1999 Sep; 17(9):

2639-2648. Colombat P, Salles G, Brousse N, Eftekhari P, Soubeyran P, Delwail V, Deconinck E, Haïoun C,

Foussard C, Sebban C, Stamatoullas A, Milpied N, Boué F, Taillan B, Lederlin P, Najman A, Czuczman MS (1999) CHOP plus rituximab chemo-immunotherapy of indolent B-cell lymphoma.

Seminars in Oncology, 1999 Oct; 26(5 Suppl 14): 88-96.

Colombat P, Salles G, Brousse N, Eftekhari P, Soubeyran P, Delwail V, Deconinck E, Haioun C,

Foussard C, Sebban C, Stamatoullas A, Milpied N, Bouè F, Taillan B, Lederlin P, Najman A,

Thièblemont C, Montestruc F, Mathieu-Boué A, Benzohra A, Solal-Céligny P (2001) Rituximab (anti-

CD20 monoclonal antibody) as single first-line therapy for patients with follicular lymphoma with a low

tumor burden: clinical and molecular evaluation. Blood, 2001 Jan 1; 97(1): 101-106.

Furge KA, Zhang Y, Woude GFV (2000) Met receptor tyrosine kinase: enhanced signaling through

adapter proteins. Oncogene, 2000 Nov; 19: 5582-5589.

Hainsworth JD (2000) Rituximab as first-line systemic therapy for patients with low-grade lymphoma.

Seminars in Oncology, 2000 Dec; 27(6 Suppl 12): 25-29.

Hainsworth JD (2002) Rituximab as first-line and maintenance therapy for patients with indolent non-

Hodgkin's lymphoma: interim follow-up of a multicenter phase II trial. Seminars in Oncology, 2002 Feb;

29(1 Suppl 2): 25-29.

Hoelzer D (2013) CD22 monoclonal antibody therapies in relapsed/refractory acute lymphoblastic

leukemia. Cancer, 2013 Aug 1; 119(15): 2671-2674.

Hudis CA (2007) Trastuzumab--mechanism of action and use in clinical practice. New England Journal

of Medicine, 2007 Jul 5; 357(1): 39-51. Kantarjian H, Thomas D, Jorgensen J, Jabbour E, Kebriaei P, Rytting M, York S, Ravandi F, Kwari M,

Faderl S, Rios MB, Cortes J, Fayad L, Tarnai R, Wang SA, Champlin R, Advani A, O'Brien S (2012)

Inotuzumab ozogamicin, an anti-CD22-calecheamicin conjugate, for refractory and relapsed acute

lymphocytic leukaemia: a phase 2 study. Lancet Oncology, 2012 Apr; 13(4): 403-411. Kantarjian H, Thomas D, Jorgensen J, Kebriaei P, Jabbour E, Rytting M, York S, Ravandi F, Garris R,

Kwari M, Faderl S, Cortes J, Champlin R, O'Brien S (2013) Results of inotuzumab ozogamicin, a CD22

monoclonal antibody, in refractory and relapsed acute lymphocytic leukemia. Cancer, 2013 Aug 1;

119(15): 2728-2736.

Lee S, Yang W, Lan KH, Sellappan S, Klos K, Hortobagyi G, Hung MC, Yu D (2002) Enhanced

sensitization to taxol-induced apoptosis by herceptin pretreatment in ErbB2-overexpressing breast cancer

cells. Cancer Research, 2002 Oct 15; 62(20): 5703-5710.

Page 24: CANCER VACCINES - Worcester Polytechnic Institute

24

Macauley MS, Crocker PR2, Paulson JC (2014) Siglec-mediated regulation of immune cell function in

disease. Nature Reviews Immunology, 2014 Oct; 14(10): 653-666.

Maloney DG, Grillo-López AJ, White CA, Bodkin D, Schilder RJ, Neidhart JA, Janakiraman N, Foon

KA, Liles TM, Dallaire BK, Wey K, Royston I, Davis T, Levy R (1997) IDEC-C2B8 (Rituximab) anti-

CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma. Blood,

1997 Sep 15; 90(6): 2188-2195. Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, Huang A, Yang NY, Nishimura M, Greve J,

Santell L, Zhang Y, Su Y, Kaufman DW, Billeci KL, Mai E, Moffat B, Lim A, Duenas ET, Phillips HS,

Xiang H, Young JC, Woude GFV, Dennis MS, Reilly DE, Schwall RH, Starovasnik MA, Lazarus RA,

Yansura DG (2013) Monovalent antibody design and mechanism of action of onartuzumab, a MET

antagonist with anti-tumor activity as a therapeutic agent. Proceedings of the National Academy of

Sciences of the United States of America, 2013 Aug; 110 (32): 2987-2996.

Naddafi F, Davami F (2015) Anti-CD19 Monoclonal Antibodies: a New Approach to Lymphoma

Therapy. International Journal of Molecular and Cellular Medicine, 2015 Summer; 4(3): 134-151.

Organ SL, Tsao MS, Bono JS (2011) An overview of of the c-MET signaling pathway. Medical

Oncology, 2011 Nov; 3(1): 7-19. Otipoby KL, Draves KE, Clark EA (2001) CD22 regulates B cell receptor-mediated signals via two

domains that independently recruit Grb2 and SHP-1. Journal of Biological Chemistry, 2001 Nov 23;

276(47): 44315-44322.

Parmiani G, De Filippo A, Novellino L, Castelli C (2007) Unique human tumor antigens: immunobiology

and use in clinical trials. Journal of Immunology, 2007 Feb 15; 178(4): 1975-1979. Perez SA, Sotiropoulou PA, Sotiriadou NN, Mamalaki A, Gritzapis AD, Echner H, Voelter W, Pawelec

G, Papamichail M, Baxevanis CN (2002) HER-2/neu-derived peptide 884-899 is expressed by human

breast, colorectal and pancreatic adenocarcinomas and is recognized by in-vitro-induced specific CD4(+)

T cell clones. Cancer Immunology, Immunotherapy, 2002 Jan; 50(11): 615-624. Piccart-Gebhart MJ1, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J,

Bell R, Jackisch C, Cameron D, et al. Herceptin Adjuvant (HERA) Trial Study Team (2005) Trastuzumab

after adjuvant chemotherapy in HER2-positive breast cancer. New England Journal of Medicine, 2005

Oct 20; 353(16): 1659-1672.

Pirker R, Pereira JR, Szczesna A, Pawel Jv, Krzakowski M, Ramlau R, Vynnychenko I, Park K, Yu CT,

Ganul V, Roh JK, Bajetta E, O’Byrne K, marinis F, Eberhardt W, Stat TGD, Emig M, Gatemeier U

(2009) Cetuximab plus chemotherapy in patients with advanced non-small-cell lung cancer (FLEX): an

open-label randomised phase III trial. The Lancet, 2009 May; 373(9674): 1497-1498. Schumacher TN, Schreiber RD (2015) Neoantigens in cancer immunotherapy. Science, 2015 Apr 3;

348(6230): 69-74. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter

J, Pegram M, Baselga J, Norton L (2001) Use of chemotherapy plus a monoclonal antibody against HER2

for metastatic breast cancer that overexpresses HER2. New England Journal of Medicine, 2001 Mar 15;

344(11): 783-792.

Page 25: CANCER VACCINES - Worcester Polytechnic Institute

25

Taylor C, Hershman D, Shah N, Suciu-Foca N, Petrylak DP, Taub R, Vahdat L, Cheng B, Pegram M,

Knutson KL, Clynes R (2007) Augmented HER-2 specific immunity during treatment with trastuzumab

and chemotherapy. Clinical Cancer Research, 2007 Sep 1; 13(17): 5133-5143. Vose JM, Link BK, Grossbard ML, Czuczman M, Grillo-Lopez A, Gilman P, Lowe A, Kunkel LA,

Fisher RI (2001) Phase II study of rituximab in combination with chop chemotherapy in patients with

previously untreated, aggressive non-Hodgkin's lymphoma. Journal of Clinical Oncology, 2001 Jan 15;

19(2): 389-397.

Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, Staehler M, Brugger W,

Dietrich PY, Mendrzyk R, Hilf N, Schoor O, Fritsche J, Mahr A, Maurer D, Vass V, Trautwein C,

Lewandrowski P, Flohr C, Pohla H, Stanczak JJ, Bronte V, Mandruzzato S, Biedermann T, Pawelec G,

Derhovanessian E, et al. (2012) Multipeptide immune response to cancer vaccine IMA901 after single-

dose cyclophosphamide associates with longer patient survival. Nature Medicine, 2012 Aug; 18(8):

1254-1261.

Bivalent Antibody Vaccines

Bi-specific (or bivalent) antibody molecules recognize two different epitopes, one for each

variable region on the antibody molecule. The rationale behind this class of cancer vaccine is that two

functions are better than one (reviewed in: Sedykh et al., 2018; Krishnamurthy and Jimeno, 2018). For

example, a bivalent antibody could recognize and bind a tumor antigen with one arm, while its other arm

recognizes and binds to an antigen on a cytotoxic T-cell. This double-binding brings the cancer cell in

close proximity to the T-cell that kills it. The binding to the T-cell antigen not only physically tethers the

cancer cell and T-cell together, in some cases (depending on the T-cell antigen) it activates the T-cell. A

study directly comparing the activities of mono-specific and bi-specific antibody treatments showed the

latter group generally has higher potency against tumor cells at a lower dosing amount and with lower

costs of production (Molhoj et al., 2007).

A convenient code for referring to bi-specific antibodies uses an “x” to separate the two binding

functions. For example, the best characterized bi-specific antibody is Blinatumomab, where one

antibody arm recognizes CD19 on the surface of B-cells (such as in leukemia), and the other antibody arm

recognizes CD3 (a T-cell activator present on the surface of T-cells). This antibody is conveniently coded

as CD19 x CD3. Other examples of bi-specific antibodies include: CD3 x glioma marker (Nitta et al.,

1990), CD3 x folate receptor (ovarian cancer cells) (Canevari et al., 1995), CD16 x CD30 (Hodgkin’s

disease) (Hartmann et al., 1997), CD319 x CD28 (B-cell lymphomas) (Daniel et al., 1998), CD64 x Fc-

Receptor (B-cell lymphomas) (Honeychurch et al., 2000), CD30 x CD64 (Hodgkin’s lymphoma)

(Borchmann et al., 2002).

Blinatumomab (CD19 x CD3) (also known as AMG-103) was the first bi-specific antibody

approved for use in the U.S. (in 2014), is (Haagen et al., 1992; Bohlen et al., 1993; Haagen et al., 1994;

DeGast et al., 1995; Weiner and DeGast, 1995). An example of Blinatumomab’s spectacular success is

seen in a 2014 clinical trial performed on 9 patients with acute lymphoblastic leukemia (ALL) (Schlegel

et al., 2014). Of the 9 patients, 4 showed complete cancer remission after one cycle of treatment, and 2

more showed complete remission after the second cycle (6 of 9 complete remissions, 67%) (Schlegel et

al., 2014). Targeting CD19 with antibodies is one of the best success stories for cancer vaccines. CD19

is present on early-stage B-cells, but it is lost when B-cells mature to plasma cells, and it is not present on

stem cells or other normal cells in the body, so these latter cells are not targeted. CD19 is an excellent

target for leukemia, because the patient produces large amounts of early-stage B-cells (hopefully

eliminated by the treatment), while the treatment would leave the stem cells and mature plasma cells

Page 26: CANCER VACCINES - Worcester Polytechnic Institute

26

(lacking CD19) alone to produce needed antibodies to fight infections (Scheuermann and Racila, 1995).

On December 3, 2014, the bi-specific antibody was approved by the FDA for treating acute lymphoblastic

leukemia (ALL) (FDA, 2014).

In non-small cell lung cancers (NSCLCs), mutations in the epidermal growth factor receptor

(EGFR) are often observed and are thought to play a role in promoting tumor growth (Harari, 2004;

Midha et al., 2015). A monoclonal antibody has been developed that targets the EGFR (Cetuximab),

though tumors can acquire resistance to these treatments through several mechanisms (Pao et al., 2005).

One mechanism identified is the over-expression of the EGFR mutant receptor c-Met, which when bound

by its ligand confers resistance (Turke et al., 2010). To overcome this limitation, scientists at Biologics

Research in Pennsylvania have created a bi-specific antibody that antagonistically binds to EGFR and

mutant c-Met, which when administered to human lung cells showed up to 80% tumor growth inhibition

(Grugan et al., 2016). Clinical trials for this bispecific antibody have not yet been reported.

Another example of a bi-specific antibody is Catumaxomab (also called Trion or Removab), the

first bi-specific and tri-functional antibody approved for use in Europe (in 2009). This antibody binds

EpCAM (present on the surface of ascites tumors) and CD3 (present on T-cells), while the Fc fragment

binds macrophages and dendritic cells (Figure-4). Catumaxomab has several methods of killing the

tumor cells, including T-cell mediated lysis, phagocytosis, and cytokine activity (secreted by the

macrophage and dendritic cells.

Figure-4: Diagram of Bispecific Antibody Catumaxomab. This

antibody binds EpCAM (upper left) present on ascites tumors, and CD3

(upper right) present on T-cells, while the Fc fragment binds

macrophage and dendritic cells (lower center). The mode of killing

includes T-cell activation, phagocytosis, and cytokine release. Figure is

from Sedykh et al., 2018.

Example Clinical Trials with Bi-specific Antibodies

Bispecific antibodies have been used since the early 1990’s in a variety of clinical trials against

several types of cancer (Table-II).

Table-II: Example Clinical Trials with Bispecific Antibodies

Targets Cancer Notes Side-Effects Reference

CD3 X CD19

B-cell non-

Hodgkin’s

lymphoma

3 human patients. The results

showed evidence of successful T-

cell activation.

Relatively safe toxicity. De Gast et

al., 1995

Page 27: CANCER VACCINES - Worcester Polytechnic Institute

27

B-cell chronic

lymphocytic

leukemia (B-

CLL)

Pre-clinical testing showed the

tumor lysis occurs mostly via

CD8+ cytotoxic T-cells, and that

CD19-negative cells (non-

leukemic cells) were not harmed.

CD19-negative cells

(non-leukemic cells)

were not harmed by the

CD8+ T-cells induced

by the antibody vaccine.

Dreier et

al., 2002

B-cell chronic

lymphocytic

leukemia (B-

CLL)

Their data showed a depletion of

lymphoma cells in 22 out of 25

(88%) patient cases, and the

depletion did not require IL-2

supplement.

Löffler et

al., 2003

CD19-positive

B-cell chronic

lymphocytic

leukemia (B-

CLL)

Used video-assisted microscopy

to show that each activated T-cell

eliminated multiple CD19 tumor

cell targets within a 9 hour time

period, and the tumor cell targets

were completely eliminated

within 24 hours using ratios as

low as 1:5.

Hoffmann

et al., 2005

Variety of

cancers

Under identical experimental

conditions, the bispecific

CD19/CD3 format has far

superior activity compared to the

monospecific formats.

Molhol et

al., 2007

Non-Hodgkin’s

lymphoma

Doses of antibody as low as

0.005 milligrams per square

meter body per day led to an

elimination of all target cells in

the blood, and partial and

complete tumor regressions were

observed at 0.015 milligram

doses. At 0.06 milligram doses,

100% of the patients

experienced tumor regression!

Bargou et

al., 2008

Acute

lymphoblastic

leukemia

(ALL)

Phase-II. 16 of 21 patients

showed a successful minimal

residual disease (MRD). 12 of

the 16 responders had been

refractory to previous cancer

treatments, so any improvement

in their condition is a significant

event.

The most frequently

observed side-effects

were grade-3 and 4

lymphopenia, but were

completely reversible.

Topp et al.,

2011

Page 28: CANCER VACCINES - Worcester Polytechnic Institute

28

Relapsing

acute

lymphoblastic

leukemia

(ALL)

Phase-II long-term follow-up of

the above study. 61% of the 20

patients had a hematologic

relapse-free survival rate. In a

subgroup of 9 patients who

progressed well enough to also

receive an allogeneic

hematopoietic stem cell

transplant, 65% showed

hematologic relapse-free

survival.

Topp et al.,

2012

B-precursor

acute

lymphoblastic

leukemia

(ALL)

Of the 9 patients, 4 achieved

complete remission after their

first cycle of treatment, 2 showed

a complete remission after the

second cycle, and the remaining 3

patients did not respond to the

treatment.

1 patient experienced

grade-3 seizures, and 2

patients experienced

grade-3 cytokine release

syndrome, but those

events were treatable.

Schlegel et

al., 2014

B-precursor

acute

lymphoblastic

leukemia (B-

ALL)

Multicenter Phase-II study. After

two treatments with CD19 x CD3

antibody, 81 of 189 patients

(43%) showed complete cancer

remission.

The most frequent

adverse events were

febrile neutropenia

(25%), neutropenia

(16%), and anemia

(14%). 2% of the

patients showed grade-3

cytokine release

syndrome. 3 deaths due

to sepsis from E. coli

and Candida were

thought to result from

the treatment by

hindering antibody

formation against the

pathogen.

Topp et al.,

2015

CD19 x

CD3

Refractory/

relapsed (r/r)

acute

lymphoblastic

leukemia

(ALL)

65 patients. The complete

remission (CR) rate was 33/65

(51%). Low responses correlated

with initial high leukemia burden

(p = .02), history of prior extra-

medullary disease (EM)

(p = .005), and active EM at the

time of treatment (p = .05). Of

the refractory cases, 41% had

evidence of EM-ALL

progression, and CD19

expression was negative (18%) or

low (23%).

Aldoss et

al., 2017

Page 29: CANCER VACCINES - Worcester Polytechnic Institute

29

CD3 x

Glioma marker

Malignant

glioma

10 patients treated with

lymphokine-activated killer

(LAK) cells treated in vitro with

bispecific antibody against CD3

(T-cell activator) x anti-glioma

marker, compared to 10 patients

treated with LAKs alone. In the

control group, 9 patients relapsed

(and 8 died within 4 years). In

the antibody-treated group no

patients relapsed (in 18 months),

4 showed tumor regression, and 4

showed tumor eradication.

Nitta et al.,

1990

CD3 x Folate

Receptor

Ovarian

carcinoma

Of the 19 patients evaluated, 3

showed complete responses

(lasting an average of 22

months), 1 showed complete

intraperitoneal response with

progressive disease in the lymph

nodes, 3 showed partial

responses, 7 had stable disease,

and 5 showed progressive

disease.

Canevari et

al., 1995

CD16 x tumor

marker CD30

Refractory

Hodgkin’s

disease

1 patient experienced complete

remission lasting 16 months, one

patient experienced partial

remission lasting 3 months, 3 had

minor responses, and 1 mixed

response.

Side effects were rare,

and consisted of fever,

lymph node pain, and a

rash.

Hartmann

et al., 1997

CD30 x

immune

activator CD64

Refractory

Hodgkin

lymphoma

Phase 1 trial. Of 10 treated

patients, 1 showed complete

remission, 3 partial remissions,

and 4 had stable disease.

The observed side

effects were transient

and mild. The most

serious were

hypotension (4 of 10),

tachycardia (6 of 10),

fatigue (10 of 10), and

fever.

Borchmann

et al., 2002

Transferrin

receptor (TfR)

x

β-secretase

(BACE1)

The team developed two

humanized bispecific antibodies

against the transferrin receptor

(TfR) (to facilitate transcytosis

across the blood brain barrier,

BBB), and against β-secretase

(BACE1) (to lower amyloid-beta

production in the brain). Dosing

primates with anti-TfR/BACE1

allowed the antibodies to cross

the BBB and reduce brain Aβ.

Wu et al.,

2014

EpCAM

(epithelial

EpCAM

positive

Antibody Catumaxomab. 16

patients, several groups of

increasing antibody doses. The

The most common

adverse events were

chills (93.8 %), fever

(87.5 %), and grade ≥3

Mau-

Sørensen et

al., 2015

Page 30: CANCER VACCINES - Worcester Polytechnic Institute

30

cancer cells) x

CD3

(T cells)

epithelial

cancer

maximum tolerated dose (MTD)

appears to be 7 µg.

increases in liver

enzymes(56.3 %). One

patient at the highest

dose of 10 µg died of

hepatic failure related to

the treatment, leading to

termination of the study.

CD30 x

CD16A (to

recruit NKs)

Heavily

pretreated

relapsed or

refractory

Hodgkin

lymphoma

Antibody AFM13, Phase-I dose-

escalation study, 28 patients.

Doses of 0.01 to 7 mg/kg body

weight. The maximum tolerated

dose was not reached. Three of

26 evaluable patients (11.5%)

achieved partial remission, and

13 patients (50%) achieved stable

disease. AFM13 was also active

in brentuximab vedotin-refractory

patients (CD30). Phase-II

currently underway.

Adverse events were

generally mild to

moderate.

Rothe et al.,

2015

CD20 x

CD3

CD20-positive

human tumors

This study addressed two of the

problems associated with

bispecific antibodies: high cost

and inconvenient

administration. They used a non-

viral DNA vector mini-circle

(MC) to produce a bispecific

antibody CD20 x CD3. The

procedure produced T-cell

mediated killing of multiple

CD20-positive tumor lines in

vitro, and delivery of the DNA to

mouse liver produced an effective

anti-cancer effect in mouse

xenograft models.

Pang et al.,

2017

EGFR x

HER3

KRAS-positive

MAPK-

positive tumors

KRAS-mutant tumors possess

abnormal MAPK pathway

signaling and cell proliferation.

This was a Phase-IB dose-

escalation study of a combination

of Cobimetinib (which blocks

MAPK signaling) and the bi-

specific antibody duligotuzumab

(which inhibits ligand binding to

two types of receptors: EGFR

and human epidermal growth

factor receptor 3 (HER3). 23

patients KRAS-mutant tumors

were enrolled. The best response

was limited to 9 patients (39%)

with stable disease.

The cobimetinib and

duligotuzumab

combination was

associated with

increased toxicity, and

limited efficacy, so the

study did not proceed to

expansion stage and

closed for enrollment.

Lieu et al.,

2017

The studies summarized above in the table illustrate that using bi-specific antibodies is better than

earlier studies with mono-specific antibodies. While the mono-specific therapies showed few complete

Page 31: CANCER VACCINES - Worcester Polytechnic Institute

31

cancer remissions, the bi-specific therapies frequently showed complete remissions, including: 22 of 25

patients (88%) (Löffler et al., 2003), 3/3 (100%) (Bargou et al., 2008), 12/20 (60%) (Topp et al., 2012),

6/9 (67%) (Schlegel et al., 2014), 81/189 (43%) (Topp et al., 2015), and 33/65 (51%) (Aldoss et al.,

2017). Thus, bispecific antibody treatments are an improvement over earlier antibody versions.

Problems with Bi-Specific Antibodies

Although bi-specific antibodies have shown strong results against cancer that are generally better

than mono-specific antibodies, they are not perfect. Bi-specific antibodies have many of the same

problems associated with mono-specific antibodies: 1) in some cases the treatment causes patient death,

2) some patient tumors become resistant to the treatment, and 3) the vaccines almost always cause side-

effects. The worst adverse effect observed was patient death. For example, one patient died from hepatic

failure caused by administration of an EpCAM x CD3 bispecific antibody treatment, leading to

termination of the entire clinical study (Mau-Sørensen et al., 2015). And in another study, 3 patients died

from E. coli and Candida sepsis caused by treatment with a CD19 x CD3 bispecific antibody that

eliminated B-cells from the patients hindering their ability to make antibodies against the pathogens

(Topp et al., 2015). But patient death was rare, and most patients treated with bispecific antibodies

showed side-effects that were generally mild, transient, and treatable (Borchmann et al., 2002; Topp et al.,

2011). With respect to patient tumors resistant to the treatments, the reason given most often in the

studies was a loss of target antigen expression by the tumor. This antigen loss was quantitated in one

study targeting CD19 in B-cell lineage tumors whose results showed that of the 41% of the non-responder

patients, CD19 expression was absent (18% of the patients) or low (23% of the patients) (Aldoss et al.,

2017). Two other problems encountered with bispecific antibodies include their high cost and their short

half-life in the body. The high cost typically results from the method of antibody production using

expensive cell culture, and the short half-life results from the rapid clearance and degradation of

antibodies passively administered to the body. Both of these latter problems have recently been addressed

using DNAs encoding the antibodies. For example, one study used a non-viral mini-circle DNA to

produce a bispecific antibody against CD20 x CD3. The procedure allowed long-term production of an

antibody that had strong anti-tumor effects in vitro against human cancer cell lines and in vivo in

humanized mouse xenograft models (Pang et al., 2017).

References for Bi-Specific Antibodies

Aldoss I, Song J, Stiller T, Nguyen T, Palmer J, O'Donnell M, Stein AS, Marcucci G, Forman S, Pullarkat

V (2017) Correlates of resistance and relapse during blinatumomab therapy for relapsed/refractory acute

lymphoblastic leukemia. American Journal of Hematology, 2017 Sep; 92(9): 858-865. doi:

10.1002/ajh.24783.

Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, Noppeney R, Viardot A, Hess G,

Schuler M, Einsele H, Brandl C, Wolf A, Kirchinger P, Klappers P, Schmidt M, Riethmüller G, Reinhardt

C, Baeuerle PA, Kufer P (2008) Tumor regression in cancer patients by very low doses of a T cell-

engaging antibody. Science, 2008 Aug 15; 321(5891): 974-977. Bohlen H, Manzke O, Patel B, Moldenhauer G, Dörken B, von Fliedner V, Diehl V, Tesch H (1993)

Cytolysis of leukemic B-cells by T-cells activated via two bispecific antibodies. Cancer Research, 1993

Sep 15; 53(18): 4310-4314. Borchmann P, Schnell R, Fuss I, Manzke O, Davis T, Lewis LD, Behnke D, Wickenhauser C, Schiller P,

Diehl V, Engert A (2002) Phase 1 trial of the novel bispecific molecule H22xKi-4 in patients with

refractory Hodgkin lymphoma. Blood, 2002 Nov 1; 100(9): 3101-3107.

Page 32: CANCER VACCINES - Worcester Polytechnic Institute

32

Canevari S, Stoter G, Arienti F, Bolis G, Colnaghi MI, Di Re EM, Eggermont AM, Goey SH, Gratama

JW, Lamers CH, et al. (1995) Regression of advanced ovarian carcinoma by intraperitoneal treatment

with autologous T lymphocytes retargeted by a bispecific monoclonal antibody. Journal of the National

Cancer Institute, 1995 Oct 4; 87(19): 1463-1469. Daniel PT, Kroidl A, Kopp J, Sturm I, Moldenhauer G, Dörken B, Pezzutto A (1998) Immunotherapy of

B-cell lymphoma with CD3x19 bispecific antibodies: costimulation via CD28 prevents "veto" apoptosis

of antibody-targeted cytotoxic T cells. Blood, 1998 Dec 15; 92(12): 4750-4757. de Gast GC, Van Houten AA, Haagen IA, Klein S, De Weger RA, Van Dijk A, Phillips J, Clark M, Bast

BJ (1995) Clinical experience with CD3 x CD19 bispecific antibodies in patients with B cell

malignancies. Journal of Hematotherapy, 1995 Oct; 4(5): 433-437. de Gast GC, Haagen IA, van Houten AA, Klein SC, Duits AJ, de Weger RA, Vroom TM, Clark MR,

Phillips J, van Dijk AJ, et al. (1995) CD8 T cell activation after intravenous administration of CD3 x

CD19 bispecific antibody in patients with non-Hodgkin lymphoma. Cancer Immunology and

Immunotherapy, 1995 Jun; 40(6): 390-396. Dreier T, Lorenczewski G, Brandl C, Hoffmann P, Syring U, Hanakam F, Kufer P, Riethmuller G,

Bargou R, Baeuerle PA (2002) Extremely potent, rapid and costimulation-independent cytotoxic T-cell

response against lymphoma cells catalyzed by a single-chain bispecific antibody. International Journal

of Cancer, 2002 Aug 20; 100(6): 690-697. FDA (2014) Blinatumomab. 3 December 2014.

http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm425597.htm

Grugan KD, Dorn K, Jarantow SW, Bushey BS, Pardinas JR, Laquerre S, Moores SL, Chiu ML (2016)

Fc-mediated activity of EGFR x c-Met bispecific antibody JNJ-61186372 enhanced killing of lung cancer

cells. mAbs, 2017 Jan; 9(1): 114-126.

Haagen IA, van de Griend R, Clark M, Geerars A, Bast B, de Gast B (1992) Killing of human

leukaemia/lymphoma B cells by activated cytotoxic T lymphocytes in the presence of a bispecific

monoclonal antibody (alpha CD3/alpha CD19). Clinical and Experimental Immunology, 1992 Dec; 90(3):

368-375. Haagen IA, Geerars AJ, de Lau WB, Clark MR, van de Griend RJ, Bast BJ, de Gast BC (1994) Killing of

autologous B-lineage malignancy using CD3 x CD19 bispecific monoclonal antibody in end stage

leukemia and lymphoma. Blood, 1994 Jul 15; 84(2): 556-563. Harari PM (2004) Epidermal growth factor receptor inhibition strategies in oncology. Endocrine-related

Cancer, 2004 Dec; 11(4): 689-708.

Hartmann F, Renner C, Jung W, Deisting C, Juwana M, Eichentopf B, Kloft M, Pfreundschuh M (1997)

Treatment of refractory Hodgkin's disease with an anti-CD16/CD30 bispecific antibody. Blood, 1997 Mar

15; 89(6): 2042-2047. Hoffmann P, Hofmeister R, Brischwein K, Brandl C, Crommer S, Bargou R, Itin C, Prang N, Baeuerle

PA (2005) Serial killing of tumor cells by cytotoxic T cells redirected with a CD19-CD3-bispecific

single-chain antibody construct. International Journal of Cancer, 2005 May 20; 115(1): 98-104. Honeychurch J, Tutt AL, Valerius T, Heijnen IA, Van De Winkel JG, Glennie MJ (2000) Therapeutic

efficacy of FcgammaRI/CD64-directed bispecific antibodies in B-cell lymphoma. Blood, 2000; 96: 3544–

3552.

Page 33: CANCER VACCINES - Worcester Polytechnic Institute

33

Krishnamurthy A, Jimeno A (2018) Bispecific antibodies for cancer therapy: A review. Pharmacology &

Therapeutics, 2018 May; 185: 122-134. doi: 10.1016/j.pharmthera.2017.12.002.

Lieu CH, Hidalgo M, Berlin JD, Ko AH, Cervantes A, LoRusso P, Gerber DE, Eder JP, Eckhardt SG,

Kapp AV, Tsuhako A, McCall B, Pirzkall A, Uyei A, Tabernero J (2017) A Phase Ib Dose-Escalation

Study of the Safety, Tolerability, and Pharmacokinetics of Cobimetinib and Duligotuzumab in Patients

with Previously Treated Locally Advanced or Metastatic Cancers with Mutant KRAS. Oncologist, 2017

Sep; 22(9): 1024-e89. doi: 10.1634/theoncologist.2017-0175.

Löffler A, Gruen M, Wuchter C, Schriever F, Kufer P, Dreier T, Hanakam F, Baeuerle PA, Bommert K,

Karawajew L, Dörken B, Bargou RC (2003) Efficient elimination of chronic lymphocytic leukaemia B

cells by autologous T cells with a bispecific anti-CD19/anti-CD3 single-chain antibody

construct. Leukemia, 2003 May; 17(5): 900-909.

Mau-Sørensen M, Dittrich C, Dienstmann R, Lassen U, Büchler W, Martinius H, Tabernero J (2015) A

phase I trial of intravenous catumaxomab: a bispecific monoclonal antibody targeting EpCAM and the T

cell coreceptor CD3. Cancer Chemotherapy and Pharmacology, 2015 May; 75(5): 1065-1073. doi:

10.1007/s00280-015-2728-5.

Midha A, Dearden S, McCormack R (2015) EGFR mutation incidence in non-small-cell lung cancer of

adenocarcinoma histology: a systematic review and global map by ethnicity. American Journal of Cancer

Research, 2015 Aug; 5(9): 2892-2911.

Mølhøj M, Crommer S, Brischwein K, Rau D, Sriskandarajah M, Hoffmann P, Kufer P, Hofmeister R,

Baeuerle PA (2007) CD19-/CD3-bispecific antibody of the BiTE class is far superior to tandem diabody

with respect to redirected tumor cell lysis. Molecular Immunology, 2007 Mar; 44(8): 1935-1943. Nitta T, Sato K, Yagita H, Okumura K, Ishii S (1990) Preliminary trial of specific targeting therapy

against malignant glioma. Lancet, 1990; 335: 368–371. [CD3 x anti-glioma marker]

Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, Kris MG, Varmus H (2005) Acquired

Resistance of Lung Adenocarcinomas to Gefitinib of Erlotinib Is Associated with a Second Mutation in

the EGFR Kinase Domain. PLoS Medicine, 2005 Feb; 2(3): e73.

Pang X, Ma F, Zhang P, Zhong Y, Zhang J, Wang T, Zheng G, Hou X, Zhao J, He C, Chen ZY (2017)

Treatment of Human B-Cell Lymphomas Using Minicircle DNA Vector Expressing Anti-CD3/CD20 in a

Mouse Model. Human Gene Therapy, 2017 Feb; 28(2): 216-225. doi: 10.1089/hum.2016.122.

Rothe A, Sasse S, Topp MS, Eichenauer DA, Hummel H, Reiners KS, Dietlein M, Kuhnert G, Kessler J,

Buerkle C, Ravic M, Knackmuss S, Marschner JP, Pogge von Strandmann E, Borchmann P, Engert A

(2015) A phase 1 study of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with

relapsed or refractory Hodgkin lymphoma. Blood, 2015 Jun 25; 125(26): 4024-4031. doi: 10.1182/blood-

2014-12-614636.

Scheuermann RH, Racila E (1995) CD19 antigen in leukemia and lymphoma diagnosis and

immunotherapy. Leukemia & Lymphoma, 1995 Aug; 18(5-6): 385-397. Schlegel P, Lang P, Zugmaier G, Ebinger M, Kreyenberg H, Witte KE, Feucht J, Pfeiffer M, Teltschik

HM, Kyzirakos C, Feuchtinger T, Handgretinger R (2014) Pediatric post-transplant relapsed/refractory B-

precursor acute lymphoblastic leukemia shows durable remission by therapy with the T-cell engaging

bispecific antibody blinatumomab. Haematologica, 2014 Jul; 99(7): 1212-1219.

Page 34: CANCER VACCINES - Worcester Polytechnic Institute

34

Sedykh SE, Prinz VV, Buneva VN, Nevinsky GA (2018) Bispecific antibodies: design, therapy,

perspectives. Drug Design, Development, and Therapy, 2018 Jan 22; 12: 195-208. doi:

10.2147/DDDT.S151282.

Topp MS, Kufer P, Gökbuget N, Goebeler M, Klinger M, Neumann S, Horst HA, Raff T, Viardot A,

Schmid M, Stelljes M, Schaich M, Degenhard E, Köhne-Volland R, Brüggemann M, Ottmann O, Pfeifer

H, Burmeister T, Nagorsen D, Schmidt M, Lutterbuese R, Reinhardt C, Baeuerle PA, Kneba M, Einsele

H, Riethmüller G, Hoelzer D, Zugmaier G, Bargou RC (2011) Targeted therapy with the T-cell-engaging

antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute

lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival.

Journal of Clinical Oncology, 2011 Jun 20; 29(18): 2493-2498. Topp MS, Gökbuget N, Zugmaier G, Degenhard E, Goebeler ME, Klinger M, Neumann SA, Horst HA,

Raff T, Viardot A, Stelljes M, Schaich M, Köhne-Volland R, Brüggemann M, Ottmann OG, Burmeister

T, Baeuerle PA, Nagorsen D, Schmidt M, Einsele H, Riethmüller G, Kneba M, Hoelzer D, Kufer P,

Bargou RC (2012) Long-term follow-up of hematologic relapse-free survival in a phase 2 study of

blinatumomab in patients with MRD in B-lineage ALL. Blood, 2012 Dec 20; 120(26): 5185-5187. Topp MS, Gökbuget N, Stein AS, Zugmaier G, O'Brien S, Bargou RC, Dombret H, Fielding AK, Heffner

L, Larson RA, Neumann S, Foà R, Litzow M, Ribera JM, Rambaldi A, Schiller G, Brüggemann M, Horst

HA, Holland C, Jia C, Maniar T, Huber B, Nagorsen D, Forman SJ, Kantarjian HM (2015) Safety and

activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic

leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncology, 2015 Jan; 16(1): 57-66.

Turke AB, Zejnullahu K, Wu YL, Song Y, Santagata DD, Lifshits E, Toschi L, Rogers A, Mok T, Sequist

L, Lindeman NI, Murphy C, Akhavanfard S, Yeap BY, Xiao Y, Capelletti M, Iafrate AJ, Lee C,

Christensen JG, Engelman JA, Janne PA (2010) Pre-existence and clonal selection of MET amplification

in EGFR mutant NSCLC. Cancer Cell, 2010 Jan; 17(1): 77-88. Weiner GJ, De Gast GC (1995) Bispecific monoclonal antibody therapy of B-cell malignancy. Leukemia

and Lymphoma, 1995 Jan; 16(3-4): 199-207. Yu YJ, Atwal JK, Zhang Y, Tong RK, Wildsmith KR, Tan C, Bien-Ly N, Hersom M, Maloney JA,

Meilandt WJ, Bumbaca D, Gadkar K, Hoyte K, Luk W, Lu Y, Ernst JA, Scearce-Levie K, Couch JA,

Dennis MS, Watts RJ (2014) Therapeutic bispecific antibodies cross the blood-brain barrier in nonhuman

primates. Science Translational Medicine, 2014 Nov 5; 6(261): 261ra154. doi:

10.1126/scitranslmed.3009835.

Antibody-Drug Conjugate Vaccines (ADCs)

In addition to monovalent and bivalent antibody vaccines, the third type of antibody vaccine is

the antibody-drug conjugate (ADC). Simply targeting and attaching an antibody by itself to a cancer

cell antigen does not always kill the cell (Thomas et al., 2016). The attachment does not always attract

sufficient attention from the immune system to remove the tagged tumor. So, scientists have designed

new types of antibody drugs, ADCs, that combine the power of antibodies (to recognize and bind specific

antigens) with cytotoxic drugs (new highly potent drugs that kill cells in very small quantities).

An ADC drug (Figure-5) typically contains an antibody directed against a tumor antigen

connected by a linker to a cytotoxic drug (that kills the cancer cell) (Casi and Neri, 2012; Flygare et al.,

Page 35: CANCER VACCINES - Worcester Polytechnic Institute

35

2012; Bouchard et al., 2014; Perez et al., 2014; Chiu and Gilliland, 2016; Kraynov et al., 2016). For a

well-designed ADC, the antibody should strongly bind a tumor-specific antigen, the antigen should not be

expressed strongly by normal cells, the linker should not release the cytotoxic drug (cargo) prematurely

into the circulation, and the toxic cargo should be potent enough to kill a cell with only a few molecules.

Figure-5: General Structure of an Antibody-Drug Conjugate. Shown is the

general structure of an ADC, consisting of a “Y”-shaped antibody (brown),

bound to a highly potent cytotoxic drug (green, diagram right) using a linker

(diagram center). Shown in text are the required properties for each component

(Thomas et al., 2016).

Most ADCs are designed to be internalized in the cancer cell. Once the ADC binds to its target

antigen, the cell engulfs the ADC by receptor-mediated endocytosis, and the ADC enters a membrane-

enclosed endocytic vesicle (endosome). This vesicle becomes acidified, which in some cases releases the

cytotoxic drug into the cytoplasm where it kills the cell (Pastan et al., 2006). To make a good ADC, the

antibody should strongly bind the antigen, the antigen should be highly expressed on the tumor cell (not

normal cells), the linker should be stable in the patient’s circulation to avoid releasing the toxin too soon,

and the cargo drug should be highly potent since only a few molecules will enter the cell.

Several types of cytotoxic drugs are used in ADCs, the most commonly used include drugs that:

1) bind DNA (leading to DNA degradation or alkylation) (i.e. calicheamicin and nemorubicin), 2) block

tubulin (inhibiting cancer cell division) (i.e. DM1 and MMAE), or 3) inhibit RNA polymerases (blocking

ancer cell RNA synthesis and gene expression) (Thomas et al., 2016). Usually 2-4 drug molecules are

attached to each antibody at locations that do not hinder interaction with the antigen (Hughes, 2010).

ADCs also have several types of linkers. The antibody can be linked to drug by one of four

methods: 1) disulfide bond formation, 2) glycol-conjugation, 3) protein tags, or 4) amino acid

incorporation (Pastan et al., 2006; Agarwal and Bertozzi, 2015; Thomas et al., 2016). Linkers can be

cleavable (degraded by proteases inside the endocytic vesicle), or non-cleavable (degraded along with the

antibody inside the vesicle) (Doronina et al., 2006; Jain et al., 2015). Cleavable linkers are usually more

stable in the bloodstream (Thomas et al., 2016). Linkers can be placed at antibody variable regions, hinge

regions, constant regions, or any combination (Agarwal and Bertozzi, 2015).

ADC Examples

Two ADCs are currently approved by the FDA: Trastuzumab emtansine (Kadcyla®) and

Brentuximab vedotin (Adcetris®) (Thomas et al., 2016). A third ADC, Gemtuzumab ozogamicin

(Mylotarg®), was initially approved but was later withdrawn (Nelson, 2010; Richwine, 2010). Kadcyla®

Page 36: CANCER VACCINES - Worcester Polytechnic Institute

36

was developed by Genentech to treat HER2-positive metastatic breast cancer in patients resistant to other

treatments (Niculescu-Duvaz, 2010; LoRusso et al., 2011; Lopus, 2011; Verma et al., 2012; Drugs.com,

2013; About Kadcyla, 2017). Kadcyla is composed of an antibody (Trastuzumab, Herceptin) against the

HER2 receptor on the surface of some types of breast cancer cells conjugated to the cytotoxic drug DM1

(Barok et al., 2014; Jain et al., 2015).

Adcetris® is used to treat CD30-positive lympho-proliferative disorders, including Hodgkin

lymphoma (HL) and anaplastic large cell lymphoma (ALCL) (Van de Donk and Dhimolea, 2012;

Brentuximab vedotin, 2016). CD30 often occurs on the surface of cells of these tumor types, but rarely on

normal cells (Küppers and Hansmann, 2005). Adcetris (Figure-6) contains an antibody (Brentuximab or

cAC10) against CD30 conjugated to 3-5 molecules of the drug monomethyl auristatin E (MMAE) using a

cathepsin-cleavable linker (Van de Donk and Dhimolea, 2012).

Figure-6: Structure of the ADC Drug Adcetris. This antibody-drug

conjugate consists of a mouse-human chimeric monoclonal antibody against

CD30 (yellow) linked via a cathepsin-cleavable linker (blue) to 3-5 units of the

cytotoxic drug monomethyl auristatin-E (MMAE) (purple) that strongly binds

tubulin and blocks cell division (Francisco et al., 2003).

More than 40 ADCs are in the clinical trial stages of development (Thomas et al., 2016), and the

future of ADCs seems bright. However, ADCs are not perfect. Sometimes an ADC drug shows strong

pre-clinical data in mice, but this strong performance does not always carry over to the human clinical

trials. In addition, all ADCs produce adverse side-effects (although they appear to be mostly manageable,

and the side-effects pale in comparison to the poor patient prognosis for untreatable cancer). And ADCs

show varying levels of effectiveness. Therefore, it is necessary to continue developing improved ADCs

that are more effective with fewer side-effects.

First-Generation Versus Second-Generation ADCs

Over the years, the design of ADC drugs has improved significantly (Thomas et al., 2016). Early

ADCs contained mouse monoclonal antibodies (mAbs) against the target antigen, but injecting a mouse

antibody into a human patient often stimulated immune rejection (or lowered half-life) of the drug

(Teicher and Chari, 2011). Early ADCs also had short half-lives in the blood, releasing their toxic

payload into the bloodstream instead of the cancer cell. And the cytotoxic payloads (such as doxorubicin,

vinblastine, or methotrexate) were not very potent, requiring the internalization of multiple ADCs per

cell.

Second-generation ADCs contain mouse-human chimera antibodies or fully humanized

antibodies that produce less of an immune rejection in the patients. An example of an ADC with a fully

Page 37: CANCER VACCINES - Worcester Polytechnic Institute

37

human antibody is CDX-011 (Keir and Vahdat, 2012). In addition, ADC linkers have been designed to be

specifically cleaved by proteases or the acidic environment inside the endocytic vesicle, releasing the

toxic cargo only inside the cancer cell. Moreover, newer ADCs use highly potent payloads (such as

Calicheamicin, Maytansine derivatives like DM1, or Auristatins like MMAE) that are far more toxic

(Lopus, 2011). These highly potent drugs have IC50 values in the nano-molar range compared to the

micro-molar range for first-generation drugs, so they have the same cell-killing effectiveness at 1000-fold

lower concentrations.

ADC Clinical Trials

ADC clinical trials have been performed on a variety of target antigens, including some of the

examples shown in Table-III.

Table-III: Example ADC Clinical Trials

Target Antigen ADC Drug Trial Type

References

CD19 ADC SAR-3419

Phase-I Younes et al., 2009 Coiffier et al., 2011

CD22 Inotuzumab ozogamicin CAT-8015

Moxetumomab pasudotox

Phase-I Advani et al., 2010 Kreitman et al., 2012

Phase-II Kantarjian et al., 2012 Wagner-Johnson et al., 2015

CD30 Brentuximab vedotin Adcetris®

Phase-I Seattle Genetics, 2010 Younes et al., 2010 Younes et al., 2013

Phase-II Younes et al., 2012 Pro et al., 2012

CD33 Gemtuzumab ozogamicin, Mylotarg® Phase-II Daver et al., 2016

Phase-III Castaigne et al., 2012 Petersdorf et al., 2013 Hills et al., 2014

Her2 Kadcyla® Trastuzumab emtansine

T-DM1

Phase-I Krop et al., 2010

Phase-II Burris et al., 2011 Perez et al., 2014 Phillips et al., 2014

Phase-III Verma et al., 2012 Krop et al., 2014

gpNMB Glembatumumab vedotin CDX-011

Phase-I Hamid et al., 2010 Bendell et al., 2014

Page 38: CANCER VACCINES - Worcester Polytechnic Institute

38

(formerly CR011-vcMMAE) Phase-II Yardley et al., 2015

Trop-2 Sacituzumab govitecan IMMU-132

Phase-I Starodub et al., 2015 Faltas et al., 2016

EGFR Depatuxizumab Mafodotin (ABT-414) Phase-I Reardon et al., 2017 Gan et al., 2018

DLL3 Rovalpituzumab Phase-I Rudin et al., 2017

The ADC clinical trials performed to date show a wide range of results, from complete remissions

to no response (Sassoon and Blanc, 2013). But some of the trials can be called spectacular successes,

including the 46% complete remissions seen with a CD22-targeting ADC (Kantarjian et al., 2012), and

the 95% complete remission (21 of 22 patients) seen for CD30-targeting Adcetris (Pro et al., 2012). Side-

effects occurred in most trials, but they were relatively mild and treatable, and should be considered a

necessary risk for these recurring untreatable cancers. Neutropenia (low neutrophil count) was almost

always observed, but it was not fatal. Other common side-effects (as with Adcetris) were: Grade 3 or 4

(serious) adverse events of neutropenia (21%), thrombocytopenia (14%), and peripheral sensory

neuropathy (12%) (Pro et al., 2012).

ADC Side-Effects

Most of the clinical trials performed with ADC drugs showed some side-effects caused by the

treatments. Examples of the side-effects are shown in Table-IV:

Table-IV: Example ADC Side-Effects

Trial

Type Reference Summary of Reported Side-Effects

Phase-I Younes et al.,

2009 Occular problems (such as blurred vision), but no other clinically significant

toxicities.

Phase-I Coiffier et al.,

2011 Ocular toxicity, but the incidence (2%) and severity were low. The hematological

toxicity was insignificant.

Phase-I Advani et al.,

2010 Common adverse effects were thrombocytopenia (decrease in platelets) (90% of

patients), asthenia (weakness) (67%), nausea (51%), and neutropenia (decrease in

neutrophils) (51%).

Phase-I Kreitman et al.,

2012 At the doses used, no dose-limiting toxicity was observed. Minor side-effects

(seen in 25-64% of the patients) included: hypo-albuminemia (low serum

albumin), aminotransferase elevations (mild liver damage), edema, headache,

hypotension, nausea, and fatigue.

Phase-II Kantarjian et

al., 2012 The most frequent adverse effects were: fever (41%), hypotension (26%), and

grade 1-2 liver problems (24%). Two patients died within 4 weeks of starting

treatment, but it was not clear whether the deaths resulted from the treatment or

the cancer.

Page 39: CANCER VACCINES - Worcester Polytechnic Institute

39

Phase-II Wagner-

Johnson et al.,

2015

Common grade 3 or 4 side-effects during the R-INO portion of the treatment

included: thrombocytopenia, lymphopenia, and neutropenia.

Phase-I Younes et al.,

2010 The most common side-effects were fatigue, pyrexia, diarrhea, nausea,

neutropenia and peripheral neuropathy.

Phase-II Younes et al.,

2012 The most common treatment-related adverse effects were peripheral neuropathy,

nausea, fatigue, neutropenia, and diarrhea.

Phase-II Pro et al., 2012 Grade 3 or 4 adverse events included neutropenia (21%), thrombocytopenia

(14%), and peripheral sensory neuropathy (12%)

Phase-I Younes et al.,

2013 Adverse events were generally grade 1 or 2, but occurred in 41% of all patients.

Phase-III Castaigne et al.,

2012 Persistent thrombocytopenia (16%).

Phase-III Petersdorf et

al., 2013 None reported.

Phase-III

Hills et al.,

2014 Doses of Mylotarg at 3 mg/m2 were associated with fewer early deaths than the

higher dose of 6 mg/m2.

Phase-II Daver et al.,

2016 The most frequent side-effects observed were nausea, mucositis, and hemorrhage.

Phase-I Krop et al.,

2010 The most common drug-related adverse events were thrombocytopenia, elevated

transaminases, fatigue, nausea, and anemia. No serious cardiac events that would

have required drug lowering were observed.

Phase-II Burris et al.,

2011 The drug appeared to be well tolerated; the most frequent side-effects were only at

grade-1 or -2 (mild). Observed grade-3 (serious) problems included hypokalemia

(lowered serum potassium levels) (8.9%), thrombocytopenia (8.0%), and fatigue

(4.5%), although these were observed only in a small minority of patients.

Phase-III Verma et al.,

2012 Grade-3 (serious) adverse events decreased from 57% to 41%. Thrombocytopenia

and liver damage were higher with Kadcyla, while diarrhea, nausea, vomiting, and

erythrodysthesia were higher with the chemotherapy.

Phase-

III Krop et al.,

2014 The Kadcyla group showed higher incidence of thrombocytopenia (5% versus

2%), but had lower incidence of neutropenia and diarrhea.

Phase-II Perez et al.,

2014 No major events reported.

Phase-II Phillips et al.,

2014 Tested a combined treatment which caused only mild grade-1 and -2 adverse

events which were treatable.

Phase-I Hamid et al.,

2010 No major events reported.

Page 40: CANCER VACCINES - Worcester Polytechnic Institute

40

Phase-I Bendell et al.,

2014 Initially, the maximum tolerated dose (MTD) was determined to be 1.34 mg/kg

(limited by the patient’s worsening neuropathy), but the MTD was increased to

1.88 mg/kg (their formal Phase-II dose) after eliminating patients with baseline

neuropathy.

Phase-II Yardley et al.,

2015 The ADC drug showed less hematologic toxicity than the chemotherapy, but

produced more rashes, pruritus (itching), neuropathy, and alopecia. The authors

concluded that the ADC was well tolerated.

Phase-

I Starodub et al.,

2015 The MTD was determined to be 12 mg/kg for one cycle of treatment, but that dose

could not be continued for additional cycles due to the formation of

neutropenia. After extended treatments at a lower dose of 10 mg/kg, no level-4

(serious) adverse events were observed, and grade-3 toxicities were fatigue,

neutropenia, diarrhea, and leukopenia.

Phase-I Faltas et al.,

2016 The drug was well tolerated.

Phase-I Rudin et al.,

2016 At a treatment amount of 0.8 mg/kg administered every three weeks, grade 4

thrombocytopenia was observed in two patients. Grade 3 adverse effects that were

noted include thrombocytopenia (11% of individuals), pleural effusion (8%), and

increased lipase (7%). The MTD noted was 0.4 mg/kg every 3 weeks.

Phase-I Reardon et al.,

2017 The most common adverse effects noted were blurred vision, dry eyes, keratitis,

photophobia, and eye pain. MTD was determined to be 2.4 mg/kg.

Phase-I Gan et al., 2018 The most common adverse effects noted were ocular related, occurring in 92% of

patients. Keratitis was the most observed adverse effect. The MTD was

determined to be 1.25 mg/kg.

ADC Problems and Future Directions

Although ADC drugs show great promise, they are not perfect. The continued approval of more

ADC drugs by the FDA will likely require continued improvements in their targeting and efficacy

(Panowski et al., 2014). Thus, there is always room for ADC improvement.

ADC drugs are complex, requiring a number of key steps to be effective. The disruption of any

of these steps can lower the effectiveness of the drug (Loganzo et al., 2016). The ADC must travel

through the circulatory system without losing its toxic cargo. It must bind the tumor cell without

targeting normal cells. It must be internalized into the cell using the correct vesicle which either degrades

the linker or degrades the entire complex, releasing the drug into the cytoplasm. The cytotoxic drug must

correctly localize to the proper cellular compartment (nucleus for DNA damaging agents, microtubules

for tubulin-binding drugs). A response by a tumor cell to alter any of these key steps can lower drug

effectiveness. Thus, there is room for improvement in each of these processes.

For example, in the situation of a tumor cell that has down-regulated the expression of the target

antigen, thus evading the ADC, the treatment strategy could be altered to include a different ADC that

targets a different antigen on the same tumor cell (Loganzo et al., 2015). Or alternatively, if a tumor cell

over time mutates its DNA to where the gene encoding tubulin expresses a product that no longer binds

DM1 or MMAE cytotoxic drugs (Kavallaris, 2010; Gillet and Gottesman, 2010; Holohan et al., 2013),

then perhaps switching to an ADC that kills by a different mechanism would help. And with respect to

Page 41: CANCER VACCINES - Worcester Polytechnic Institute

41

drug movement from the vesicle into the cytoplasm, in some cases the drug is carried outside the cell by

drug transporter molecules such as MDR1 and MRP1, decreasing drug effectiveness (Chen et al., 2015;

Yu et al., 2015). Perhaps this problem could be overcome by co-treating with a drug to lower MDR1 or

MRP1 expression.

The following topics were identified in our review of the ADC literature as potential future

directions:

1) Cargo Switching: In some cases, a patient’s tumor can become resistant to the cytotoxic drug used in

a therapy, so perhaps switching to a different ADC that targets the same antigen but contains a different

cytotoxic cargo that works by a different mechanism might improve effectiveness. This switching

approach has successfully been used in a mouse model of non-Hodgkin lymphoma (NHL), where altering

the CD22-targeting ADC payload from MMAE (which blocks tubulin polymerization, preventing cell

division) to nemorubicin (which targets DNA) overcame the resistance (Yu et al., 2015).

2) Closely Monitoring Target Antigen Expression: One of the best practices observed in the clinical

trials was the constant monitoring of target antigen expression by the patient’s tumor cells. In some

cases, the tumor down-regulates antigen expression, so any targeted therapy (such as an ADC) no longer

targets those cells. The best clinical response rates were observed for patients still expressing the target

antigen. Antigen expression can be monitored by IHC (immuno-histochemistry), or RT-PCR (reverse

transcriptase polymerase chain reaction).

3) Dual-Targeted Therapies: This approach uses two different ADCs targeting different antigens on the

same tumor. This strategy would allow continued targeting, even in those cases where the tumor cells no

longer express one of the target antigens.

4) New ADC Conjugation Reactions: Chemical conjugation reactions are used to link the antibody to

the drug cargo. Early-generation ADCs used conjugation reactions that could add drug molecules onto

any site on the antibody containing a reactive amino acid (Panowski et al., 2014; Agarwal et al.,

2015). But these reactions added the drugs randomly onto the antibody, producing a heterogeneous

mixture of ADC molecules, each with their own activities. And each production batch varied in

composition, making it difficult to compare clinical trial data (Panowski et al., 2014; Agarwal et al.,

2015). Newer methods of conjugation allow the drug to be added to specific sites, helping eliminate

heterogeneity. The newer methods include the use of engineered cysteine residues, and the use of non-

natural amino acids.

ADC References Cited

About Kadcyla (2017) Genentech. Retrieved from: http://www.kadcyla.com/hcp/drug-

information/mechanism-of-action. Advani A, Coiffier B, Czuczman MS, Dreyling M, Foran J, Gine E, Gisselbrecht C, Ketterer N, Nasta S,

Rohatiner A, Schmidt-Wolf IG, Schuler M, Sierra J, Smith MR, Verhoef G, Winter JN, Boni J,

Vandendries E, Shapiro M, Fayad L (2010) Safety, Pharmacokinetics, And Preliminary Clinical Activity

of Inotuzumab Ozogamicin, A Novel Immunoconjugate for the Treatment of B-Cell Non-Hodgkin's

Lymphoma: Results of a Phase I Study. Journal of Clinical Oncology, 2010 Apr 20; 28(12): 2085-2093. Agarwal P, Bertozzi CR (2015) Site-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal

Chemistry, Protein Engineering, and Drug Development. Bioconjugate Chemistry, 2015 Feb 18; 26(2):

176-192.

Page 42: CANCER VACCINES - Worcester Polytechnic Institute

42

Barok M, Joensuu H, Isola J (2014) Trastuzumab Emtansine: Mechanisms of Action and Resistance.

Breast Cancer Research, 16: 209.

Bendell J, Saleh M, Rose AA, Siegel PM, Hart L, Sirpal S, Jones S, Green J, Crowley E, Simantov R,

Keler T, Davis T, Vahdat L (2014) Phase I/II Study of the Antibody-Drug Conjugate Glembatumumab

Vedotin in Patients With Locally Advanced or Metastatic Breast Cancer. Journal of Clinical Oncology,

2014 Nov 10; 32(32): 3619-3625. Bouchard H, Viskov C, Garcia-Echeverria C (2014) Antibody-Drug Conjugates: A New Wave of Cancer

Drugs. Bioorganic and Medicinal Chemistry Letters, 24 (23): 5357-5363. Brentuximab vedotin (2016) Drugbank. https://www.drugbank.ca/drugs/DB08870

Burris H, Saleh M, Bendell J, Hart L, Rose A, Dong Z, Siegel P, Crane M, Donovan D, Crowley D,

Simantov R, Vahdat L (2009) A Phase (Ph) I/II Study of CR011-VcMMAE, an Antibody-Drug

Conjugate, in Patients (Pts) with Locally Advanced or Metastatic Breast Cancer (MBC). Cancer

Research, 69 (Meeting Abstract Supplement): 6096. Burris HA, Rugo HS, Vukelja SJ, Vogel CL, Borson RA, Limentani S, Tan-Chiu E, Krop IE, Michaelson

RA, Girish S, Amler L, Zheng M, Chu YW, Klencke B, O'Shaughnessy JA (2011) Phase II Study of the

Antibody Drug Conjugate Trastuzumab-DM1 for the Treatment of Human Epidermal Growth Factor

Receptor 2 (HER2)-Positive Breast Cancer After Prior HER2-Directed Therapy. Journal of Clinical

Oncology, 2011 Feb 1; 29(4): 398-405.

Casi G, Neri D (2012) Antibody–Drug Conjugates: Basic Concepts, Examples and

Future Perspectives. Journal of Controlled Release, 161(2): 422-428.

Castaigne S, Pautas C, Terré C, Raffoux E, Bordessoule D, Bastie JN, Legrand O, Thomas X, Turlure P,

Reman O, de Revel T, Gastaud L, de Gunzburg N, Contentin N, Henry E, Marolleau JP, Aljijakli A,

Rousselot P, Fenaux P, Preudhomme C, Chevret S, Dombret H (2012) Acute Leukemia French

Association. Effect of Gemtuzumab Ozogamicin on Survival of Adult Patients With De-Novo Acute

Myeloid Leukaemia (ALFA-0701): A Randomised, Open-Label, Phase 3 Study. Lancet, 2012 Apr 21;

379(9825): 1508-1516.

Chen R, Hou J, Newman E, Kim Y, Donohue C, Liu X, Thomas SH, Forman SJ, Kane SE

(2015) CD30 Downregulation, MMAE Resistance, and MDR1 Upregulation Are All Associated

with Resistance to Brentuximab Vedotin. Molecular Cancer Therapeutics, 2015 Jun; 14(6):

1376-1384.

Chiu ML, Gilliland GL (2016) Engineering Antibody Therapeutics. Structural Biology, 38: 163-173. Coiffier B, Ribrag V, Dupuis J, Tilly H, Haioun C, Morschhauseret F, et al. (2011) Phase I/II Study of the

Anti-CD19 Maytansinoid Immunoconjugate SAR3419 Administered Weekly to Patients With

Relapsed/Refractory B-Cell Non-Hodgkin's Lymphoma (NHL). Journal of Clinical Oncology, 2011; 29

(Supplement): 8017.

Daver N, Kantarjian H, Ravandi F, Estey E, Wang X, Garcia-Manero G, Jabbour E, Konopleva M,

O'Brien S, Verstovsek S, Kadia T, Dinardo C, Pierce S, Huang X, Pemmaraju N, Diaz-Pines-Mateo M,

Cortes J, Borthakur G (2016) A Phase II Study of Decitabine and Gemtuzumab Ozogamicin in Newly

Diagnosed and Relapsed Acute Myeloid Leukemia and High-Risk Myelodysplastic Syndrome. Leukemia,

2016 Feb; 30(2): 268-273.

Page 43: CANCER VACCINES - Worcester Polytechnic Institute

43

Doronina SO, Mendelsohn BA, Bovee TD, Cerveny CG, Alley SC, Meyer DL, Oflazoglu E,

Toki BE, Sanderson RJ, Zabinski RF, Wahl AF, Senter PD (2006) Enhanced Activity of

Monomethylauristatin F Through Monoclonal Antibody Delivery: Effects of Linker Technology

on Efficacy and Toxicity. Bioconjugate Chemistry, 2006 Jan-Feb; 17(1): 114-124.

Drugs.com (2013) FDA Approves Kadcyla. February 22, 2013. Elias DJ, Hirschowitz L, Kline LE, Kroener JF, Dillman RO, Walker LE, Robb JA, Timms RM (1990)

Phase I Clinical Comparative Study of Monoclonal Antibody KS1/4 and KS1/4-Methotrexate

Immunconjugate in Patients With Non-Small Cell Lung Carcinoma. Cancer Research, 1990 Jul 1;

50(13): 4154-4159. Faltas B, Goldenberg DM, Ocean AJ, Govindan SV, Wilhelm F, Sharkey RM, Hajdenberg J, Hodes G,

Nanus DM, Tagawa ST (2016) Sacituzumab Govitecan, a Novel Antibody-Drug Conjugate, in Patients

With Metastatic Platinum-Resistant Urothelial Carcinoma. Clinical Genitourinary Cancer, 2016 Feb;

14(1): e75-79.

Francisco JA, Cerveny CG, Meyer DL, Mixan BJ, Klussman K, Chace DF, Rejniak SX, Gordon KA,

DeBlanc R, Toki BE, Law CL, Doronina SO, Siegall CB, Senter PD, Wahl AF (2003) cAC10-vcMMAE,

An Anti-CD30-Monomethyl Auristatin E Conjugate with Potent and Selective Antitumor Activity. Blood,

2003 Aug 15; 102(4): 1458-1465.

Flygare JA, Pillow TH, & Aristoff P (2012) Antibody-Drug Conjugates for the Treatment of

Cancer. Chemical Biology & Drug Design, 81(1): 113-121.

Gan HK, Reardon DA, Lassman AB, Merrell R, Bent VDM, Butowski N, Lwin Z, Wheeler H, Fichtel L,

Scott AM, Gomez EJ, Fischer J, Mandich H, Xiong H, Lee HJ, Munasinghe WP, Roberts-Rapp LA,

Ansell PJ, Holen KD, Kumthekar P (2018) Safety, pharmacokinetics, and antitumor response of

depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with

glioblastoma. Neuro Oncology, 2018 May 18; 20(6): 838-847. Gemtuzumab Ozogamicin (Mylotarg®) Drug Description (2015) ADC Reviews

https://adcreview.com/gemtuzumab-ozogamicin-mylotarg/ Gillet JP, Gottesman MM (2010) Mechanisms of Multidrug Resistance in Cancer. Methods in Molecular

Biology, 2010; 596: 47-76.

Hamid O, Sznol M, Pavlick AC, et al. (2010) Frequent Dosing And GPNMB Expression With CDX-011

(CR011-Vcmmae), An Antibody-Drug Conjugate (ADC), In Patients With Advanced Melanoma. Journal

of Clinical Oncology, 28: 15s, 2010 (Supplement; Abstract 8525).

Hughes B (2010) Antibody–Drug Conjugates for Cancer: Poised to Deliver? Nature Reviews Drug

Discovery, 9: 665-667.

Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M, Estey EH, Dombret H,

Chevret S, Ifrah N, Cahn JY, Récher C, Chilton L, Moorman AV, Burnett AK (2014) Addition of

Gemtuzumab Ozogamicin to Induction Chemotherapy in Adult Patients With Acute Myeloid Leukaemia:

A Meta-Analysis of Individual Patient Data From Randomised Controlled Trials. The Lancet, Oncology,

2014 Aug; 15(9): 986-996.

Page 44: CANCER VACCINES - Worcester Polytechnic Institute

44

Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG (2013) Cancer Drug Resistance: An

Evolving Paradigm. Nature Reviews, Cancer, 2013 Oct; 13(10): 714-726.

Jain N, Smith SW, Ghone S, Tomczuk B (2015) Current ADC Linker Chemistry. Pharmaceutical

Research, 32(11): 3526-3540.

Kantarjian H, Thomas D, Jorgensen J, Jabbour E, Kebriaei P, Rytting M, York S, Ravandi F, Kwari M,

Faderl S, Rios MB, Cortes J, Fayad L, Tarnai R, Wang SA, Champlin R, Advani A, O'Brien S (2012)

Inotuzumab Ozogamicin, An Anti-CD22-Calecheamicin Conjugate, For Refractory and Relapsed Acute

Lymphocytic Leukaemia: A Phase 2 Study. The Lancet, Oncology, 2012 April; 13(4): 403-411.

Kavallaris M (2010) Microtubules and Resistance to Tubulin-Binding Agents. Nature Reviews,

Cancer, 2010 Mar; 10(3): 194-204.

Keir CH, Vahdat LT (2012) The Use of an Antibody Drug Conjugate, Glembatumumab Vedotin (CDX-

011), For the Treatment of Breast Cancer. Expert Opinion on Biological Therapy, 2012 Feb; 12(2): 259-

263.

Kraynov E, Kamath AV, Walles M, Tarcsa E, Deslandes A, Iyer RA, Datta-Mannan A, Sriraman P,

Bairlein M, Yang JJ, Barfield M, Xiao G, Escandon E, Wang W, Rock DA, Chemuturi NV, Moore DJ

(2016) Current Approaches for Absorption, Distribution, Metabolism, and Excretion Characterization of

Antibody-Drug Conjugates: An Industry White Paper. Drug Metabolism and Disposition, 2016 May;

44(5): 617-623.

Kreitman RJ, Tallman MS, Robak T, Coutre S, Wilson WH, Stetler-Stevenson M, Fitzgerald DJ,

Lechleider R, Pastan I (2012) Phase I Trial of Anti-CD22 Recombinant Immunotoxin Moxetumomab

Pasudotox (CAT-8015 Or HA22) in Patients With Hairy Cell Leukemia. Journal of Clinical Oncology,

2012 May 20; 30(15): 1822-1828.

Krop IE, Beeram M, Modi S, Jones SF, Holden SN, Yu W, Girish S, Tibbitts J, Yi JH, Sliwkowski MX,

Jacobson F, Lutzker SG, Burris HA (2010) Phase I Study of Trastuzumab-DM1, An HER2 Antibody-

Drug Conjugate, Given Every 3 Weeks to Patients With HER2-Positive Metastatic Breast Cancer.

Journal of Clinical Oncology, 2010 Jun 1; 28(16): 2698-2704.

Krop IE, Kim SB, González-Martín A, LoRusso PM, Ferrero JM, Smitt M, Yu R, Leung AC, Wildiers H;

TH3RESA study collaborators (2014) Trastuzumab Emtansine Versus Treatment of Physician's Choice

for Pretreated HER2-Positive Advanced Breast Cancer (TH3RESA): A Randomised, Open-Label, Phase

3 Trial. Lancet Oncology, 2014 Jun;15(7): 689-699.

Küppers R, Hansmann ML (2005) The Hodgkin and Reed/Sternberg Cell. International Journal of

Biochemistry and Cell Biology, 2005 Mar; 37(3): 511-517.

Loganzo F, Tan X, Sung M, Jin G, Myers JS, Melamud E, Wang F, Diesl V, Follettie MT, Musto S, Lam

MH, Hu W, Charati MB, Khandke K, Kim KS, Cinque M, Lucas J, Graziani E, Maderna A, O'Donnell

CJ, Arndt KT, Gerber HP (2015a) Tumor Cells Chronically Treated With a Trastuzumab-Maytansinoid

Antibody-Drug Conjugate Develop Varied Resistance Mechanisms But Respond to Alternate Treatments.

Molecular Cancer Therapeutics, 2015 Apr; 14(4): 952-963.

Loganzo F, Sung M, Tan X, Gerber HP (2015b) Emerging Mechanisms of Resistance to Antibody Drug

Conjugates in Preclinical Models. Molecular Med Tri-Con; 2015 Feb 18; San Francisco, CA: Molecular

Med Tri-Con.

Page 45: CANCER VACCINES - Worcester Polytechnic Institute

45

Loganzo F, Tan X, Sung M, Jin G, Myers JS, Melamud E, Wang F, Diesl V, Follettie MT, Musto S, Lam

MH, Hu W, Charati MB, Khandke K, Kim KS, Cinque M, Lucas J, Graziani E, Maderna A, O'Donnell

CJ, Arndt KT, Gerber HP (2015c) Tumor Cells Chronically Treated With a Trastuzumab-Maytansinoid

Antibody-Drug Conjugate Develop Varied Resistance Mechanisms But Respond to Alternate Treatments.

Molecular Cancer Therapeutics, 2015 Apr; 14(4): 952-963.

Loganzo F, Sung M, Gerber HP (2016) Mechanisms of Resistance to Antibody-Drug Conjugates.

Molecular Cancer Therapeutics, 2016 Dec; 15(12): 2825-2834.

Lopus M (2011) Antibody-DM1 Conjugates as Cancer Therapeutics. Cancer Letters, 2011 Aug 28;

307(2): 113-118.

LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX (2011) Trastuzumab Emtansine: A Unique

Antibody-Drug Conjugate in Development for Human Epidermal Growth Factor Receptor 2-Positive

Cancer. Clinical Cancer Research, 2011 Oct 15; 17(20): 6437-6447. Nelson R (2010) Gemtuzumab Voluntarily Withdrawn from US Market. Medscape, Issue: June 21, 2010.

Niculescu-Duvaz I (2010) Trastuzumab Emtansine, An Antibody-Drug Conjugate for the Treatment of

HER2+ Metastatic Breast Cancer. Current Opinion in Molecular Therapeutics, 2010 Jun; 12(3): 350-

360.

Panowski S, Bhakta S, Raab H, Polakis P, Junutula JR (2014) Site-Specific Antibody Drug Conjugates

For Cancer Therapy. MAbs, 2014 Jan-Feb; 6(1): 34-45.

Pastan I, Hassan R, FitzGerald DJ, Kreitman RJ (2006) Immunotoxin Therapy of Cancer. Nature Reviews

Cancer, 6: 559-565. Perez EA, Hurvitz SA, Amler LC, Mundt KE, Ng V, Guardino E, Gianni L (2014) Relationship Between

HER2 Expression and Efficacy With First-Line Trastuzumab Emtansine Compared With Trastuzumab

Plus Docetaxel in TDM4450g: A Randomized Phase II Study of Patients With Previously Untreated

HER2-Positive Metastatic Breast Cancer. Breast Cancer Research, 2014 May 23; 16(3): R50.

Perez HL, Cardarelli PM, Deshpande S, Gangwar S, Schroeder GM, Vite GD, Borzilleri RM (2014)

Antibody–Drug Conjugates: Current Status and Future Directions. Drug Discovery Today, 19(7): 869-

881.

Petersdorf SH, Kopecky KJ, Slovak M, Willman C, Nevill T, Brandwein J, Larson RA, Erba HP, Stiff PJ,

Stuart RK, Walter RB, Tallman MS, Stenke L, Appelbaum FR (2013) A Phase 3 Study of Gemtuzumab

Ozogamicin During Induction and Post-Consolidation Therapy in Younger Patients With Acute Myeloid

Leukemia. Blood, 2013 Jun 13; 121(24): 4854-4860.

Phillips GD, Fields CT, Li G, Dowbenko D, Schaefer G, Miller K, Andre F, Burris HA 3rd, Albain KS,

Harbeck N, Dieras V, Crivellari D, Fang L, Guardino E, Olsen SR, Crocker LM, Sliwkowski MX (2014)

Dual Targeting of HER2-Positive Cancer With Trastuzumab Emtansine and Pertuzumab: Critical Role

For Neuregulin Blockade in Antitumor Response to Combination Therapy. Clinical Cancer Research,

2014 Jan 15; 20(2): 456-468.

Pollack A (2013) FDA Approves a New Drug for Advanced Breast Cancer. The New York Times,

February 22, 2013. http://www.nytimes.com/2013/02/23/business/fda-approves-breast-cancer-drug.html

Page 46: CANCER VACCINES - Worcester Polytechnic Institute

46

Pro B, Advani R, Brice P, Bartlett NL, Rosenblatt JD, Illidge T, Matous J, Ramchandren R, Fanale M,

Connors JM, Yang Y, Sievers EL, Kennedy DA, Shustov A (2012) Brentuximab Vedotin (SGN-35) In

Patients With Relapsed Or Refractory Systemic Anaplastic Large-Cell Lymphoma: Results of a Phase II

Study. Journal of Clinical Oncology, 2012 Jun 20; 30(18): 2190-2196.

Reardon DA, Lassman AB, Bend VDM, Kumthekar P, Merrell R, Scott AM, Fichtel L, Sulman EP,

Gomez E, Fischer J, Lee HJ, Munasinghe W, Xiong H, Mandich H, Roberts-Rapp L, Ansell P, Holen

KD, Gan HK (2017) Efficacy and safety results of ABT-414 in combination with radiation and

temozolomide in newly diagnosed glioblastoma. Neuro Oncology, 2017 Jul 1; 19(7): 965-975.

Rudin CM, Pietanza MC, Bauer ™, Ready N, Morgenstern D, Glisson BS, Byers LA, Johnson ML,

Burris HA, Robert F, Han TH, Bheddah S, Theiss N, Watson S, Mathur D, Vennapusa B, Zayed H, Lally

S, Strickland DK, Govindan R, Dylla SJ, Peng SL, Spigel DR (2017) Rovalpituzumab tesirine, a DLL3-

targeted antibody-drug conjugate, in recurrent small-cell lung cancer: a first-in-human, first-in-class,

phase 1 study. Lancet Oncology, 2017 Jan; 18(1): 42-51.

Richwine L (2010) Pfizer Pulls Leukemia Drug from U.S. Market. Reuters, June 21, 2010.

Sassoon I, Blanc V (2013) Antibody-Drug Conjugate (ADC) Clinical Pipeline: A Review. Methods in

Molecular Biology, 2013; 1045: 1-27. Seattle Genetics Press Release (2010) Seattle Genetics and Millennium Report Positive Data from Pivotal

Trial of Brentuximab Vedotin (SGN-35) in Relapsed or Refractory Hodgkin Lymphoma at ASH Annual

Meeting. December 6, 2010. http://investor.seagen.com/phoenix.zhtml?c=124860&p=irol-

newsArticle&ID=1504306&highlight= Starodub AN, Ocean AJ, Shah MA, Guarino MJ, Picozzi VJ Jr, Vahdat LT, Thomas SS, Govindan SV,

Maliakal PP, Wegener WA, Hamburger SA, Sharkey RM, Goldenberg DM (2015) First-in-Human Trial

of a Novel Anti-Trop-2 Antibody-SN-38 Conjugate, Sacituzumab Govitecan, for the Treatment of

Diverse Metastatic Solid Tumors. Clinical Cancer Research, 2015 Sep 1; 21(17): 3870-3878.

Teicher BA, Chari RV (2011) Antibody Conjugate Therapeutics: Challenges and Potential. Clinical

Cancer Research,17(20): 6389-6397.

Thomas A, Teicher BA, Hassan R (2016) Antibody-Drug Conjugates for Cancer Therapy. Lancet

Oncology, 17(6): 254-262.

Tolcher AW, Sugarman S, Gelmon KA, Cohen R, Saleh M, Isaacs C, Young L, Healey D, Onetto N,

Slichenmyer W (1999) Randomized Phase II Study of BR96-Doxorubicin Conjugate in Patients With

Metastatic Breast Cancer. Journal of Clinical Oncology, 1999 Feb; 17(2): 478-484.

Vaklavas C, Forero-Torres A (2012) Safety and Efficacy of Brentuximab Vedotin in Patients with

Hodgkin Lymphoma or Systemic Anaplastic Large Cell Lymphoma. Therapeutic Advances in

Hematology, 2012 Aug; 3(4): 209-225.

Van de Donk NW, Dhimolea E (2012) Brentuximab Vedotin. mAbs, 4(4): 458–465.

Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY, Diéras V, Guardino E,

Fang L, Lu MW, Olsen S, Blackwell K; EMILIA Study Group (2012) Trastuzumab Emtansine for HER2-

Positive Advanced Breast Cancer. New England Journal of Medicine, 2012 Nov 8; 367(19): 1783-1791.

Page 47: CANCER VACCINES - Worcester Polytechnic Institute

47

Wagner-Johnston ND, Goy A, Rodriguez MA, Ehmann WC, Hamlin PA, Radford J, Thieblemont C, Suh

C, Sweetenham J, Huang Y, Sullivan ST, Vandendries ER, Gisselbrecht C (2015) A Phase 2 Study of

Inotuzumab Ozogamicin and Rituximab, Followed by Autologous Stem Cell Transplant in Patients With

Relapsed/Refractory Diffuse Large B-Cell Lymphoma. Leukemia & Lymphoma, 2015; 56(10): 2863-

2869.

Wayne AS, Bhojwani D, Silverman LB, Richards K, Stetler-Stevenson M, Shah NN, et al. (2011) A

Novel Anti-CD22 Immunotoxin, Moxetumomab Pasudotox: Phase I Study in Pediatric Acute

Lymphoblastic Leukemia (ALL). Blood (ASH Annual Meeting Abstract), 2011; 118: 248.

Yardley DA, Weaver R, Melisko ME, Saleh MN, Arena FP, Forero A, Cigler T, Stopeck A, Citrin D,

Oliff I, Bechhold R, Loutfi R, Garcia AA, Cruickshank S, Crowley E, Green J, Hawthorne T, Yellin MJ,

Davis TA, Vahdat LT (2015) EMERGE: A Randomized Phase II Study of the Antibody-Drug Conjugate

Glembatumumab Vedotin in Advanced Glycoprotein NMB-Expressing Breast Cancer. Journal of

Clinical Oncology, 2015 May 10; 33(14): 1609-1619.

Younes A, Gordon L, Kim S, Romaguera J, Copeland AR, de Castro Farial S, et al. (2009) Phase I Multi-

Dose Escalation Study of the Anti-CD19 Maytansinoid Immunoconjugate SAR3419 Administered By

Intravenous (IV) Infusion Every 3 Weeks to Patients With Relapsed/Refractory B-Cell Non-Hodgkin's

Lymphoma (NHL). Blood, (ASH Annual Meeting Abstracts), 2009; 114: 585.

Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, Forero-Torres A (2010)

Brentuximab Vedotin (SGN-35) for Relapsed CD30-Positive Lymphomas. New England Journal of

Medicine, 2010 Nov 4; 363(19): 1812-1821.

Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, Ramchandren R, Bartlett NL,

Cheson BD, de Vos S, Forero-Torres A, Moskowitz CH, Connors JM, Engert A, Larsen EK, Kennedy

DA, Sievers EL, Chen R (2012) Results of a Pivotal Phase II Study of Brentuximab Vedotin for Patients

With Relapsed or Refractory Hodgkin's Lymphoma. Journal of Clinical Oncology, 2012 Jun 20; 30(18):

2183-2189.

Younes A, Connors JM, Park SI, Fanale M, O'Meara MM, Hunder NN, Huebner D, Ansell SM (2013)

Brentuximab Vedotin Combined With ABVD or AVD for Patients With Newly Diagnosed Hodgkin's

Lymphoma: A Phase 1, Open-Label, Dose-Escalation Study. The Lancet, Oncology, 2013 Dec; 14(13):

1348-1356.

Yu SF, Zheng B, Go M, Lau J, Spencer S, Raab H, Soriano R, Jhunjhunwala S, Cohen R, Caruso M,

Polakis P, Flygare J, Polson AG (2015) A Novel Anti-CD22 Anthracycline-Based Antibody-Drug

Conjugate (ADC) That Overcomes Resistance to Auristatin-Based ADCs. Clinical Cancer Research,

2015 Jul 15; 21(14): 3298-3306.

Page 48: CANCER VACCINES - Worcester Polytechnic Institute

48

Lit Review Section-2:

Dendritic Cell Vaccines

Leila Camplese

Half of the 2011 Nobel Prize in Physiology or Medicine went to Ralph M. Steinman for his

“discovery of the dendritic cell and its role in adaptive immunity" (Nobel Prize, 2011). As mentioned

previously in the Introduction to Immunology section, dendritic cells (DCs) are “professional” antigen-

presenting cells that reside in tissues that contact the external environment (skin, lining of the respiratory

tract, nasal epithelium, etc.). Their main function is to recognize foreign antigens on the surface of

invading pathogens, process the antigen within the DC, present the antigen on the cell surface, then

migrate to the lymph nodes to present the antigen to other cells of the immune system (T-cells and B-

cells) (Steinman and Cohn, 1973; Banchereau and Steinman, 1998; Sallusto and Lanzavecchia, 2002;

Trombetta and Mellman, 2005). These latter immune cells then differentiate and commit to that antigen

to help eliminate the threat.

In the case of cancer, the tumor cells themselves are poor antigen-presenting cells, so DCs help

facilitate the tumor removal by presenting their antigens to the immune system to induce a response to the

tumor. Animal experiments have shown that DCs are a required component of the body’s immune attack

against cancer. When a tumor forms in the body, DCs help process the tumor’s neo-antigens and present

them to the immune system to generate active B-cell and T-cell responses against the tumor (Palucka and

Banchereau, 2012).

With respect to tumor vaccines, the antigen-presenting properties of DCs are sometimes used to

“prime” a patient’s DCs against a single antigen, or mixture of antigens (Figure-7). In a common ex vivo

approach, a patient’s DC cells are isolated from peripheral blood mononuclear cells (PBMCs) using

various techniques, and are cultured to expand their numbers. The isolated DCs are then “pulsed” or

“primed” (mixed) with foreign tumor antigen (purified antigen or entire tumor cells themselves) (dagram

upper left), and the pulsed DCs are injected back into the patient. Hopefully, these primed DCs migrate

to the lymph nodes to engage B-cells and T-cells (diagram right) to commit them against the tumor

antigen (Davis et al., 2003; Steinman and Banchereau, 2007; Koski et al., 2008; Schuler, 2010; Ueno et

al., 2010). In a less used in vivo approach, DCs in the patient’s body are induced to take up tumor-specific

antigens, and the antigen-presentation is done naturally to stimulate the patient’s T-cells. This latter

process does not involve purification of the DCs nor their ex vivo expansion.

Figure-7: Diagram of a Typical DC Vaccine. In this

approach, dendritic cells (DCs) (purple) are primed with tumor

antigens (diagram upper left). The priming can be performed in

vitro or in vivo. The activated DC cells migrate to nearby

lymph nodes and present the antigens bound to MHCs (light

brown). MHC-I presents to T-cell receptors (blue) on CD8+

cells (yellow), while MHC-II presents to T-cell receptors on

CD4+ cells (also yellow). CD4+ helper T-cells produce

cytokines that promote CD8+ T-cell maturation. CD8+

cytotoxic T-cells leave the lymph node into the circulation

where they recognize the tumor cells expressing the antigen,

killing the cell. Figure from Anagnostou and Brahmer, 2015.

Page 49: CANCER VACCINES - Worcester Polytechnic Institute

49

Example Studies with DC Vaccines

The first DC vaccine approved by the FDA was Provenge (Ledford, 2015). This vaccine is also

called Sipuleucel-T or APC8015. Provenge is directed against prostate cancer, the second leading cause

of cancer death in men following skin cancer. Researchers chose prostate cancer, because so many men

have it, and also because men can live without a prostate so if the vaccine accidently targeted normal

prostate cells it would not be fatal (Ledford, 2015).

Much research went into the development of this vaccine. A patient’s peripheral blood

mononuclear cells (PBMCs) (including DCs) are isolated by leukapheresis, and cultured to increase their

numbers. They are then mixed in vitro with a patented recombinant fusion protein (PA-2024). PA-2024

used to prime the DCs contains the target antigen prostatic acid phosphatase (PAP) fused with

granulocyte macrophage colony stimulating factor (GM-CSF) which helps activate the immune system.

The primed DCs are then injected back into the same patient.

The clinical results of Provenge (Table-V) are mixed, showing some successes, followed by an

underwhelming response. Early experiments demonstrated that CTLs had been formed against the tumor

cells, and that PSA levels dropped, but the survival statistics were relatively unimpressive compared to

controls, for example increasing from 4.9 months survival before treatment to 7.9 months after treatment

(Beinart et al., 2005), but few of the clinical trials showed complete remissions. Despite Provenge’s FDA

approval, the company who developed it (Dendreon Corp.) went bankrupt. Dendreon was hurt by the

long wait for FDA approval for an early cancer vaccine (18 years), by confusion over Medicare

reimbursements, and by lukewarm results (Ledford, 2015). In 2015, the rights to Provenge were

purchased by Valeant Pharmaceuticals.

As with any clinical treatment, there are several risks associated with Provenge. Many of them

are clearly listed on their website as acute infusion reactions, including (but not limited to): fever,

headache, nausea, and joint pain (Dendreon Pharmaceuticals, 2017). More rarely the patients can

experience more severe side-effects, such as chest pain, vomiting, stroke, and thrombosis. 1.5% of the

participating patients backed out of the clinical trial upon experiencing these side effects, however this

number is so low that it would not have a significant impact on the study results (Dendreon

Pharmaceuticals, 2017).

Melanoma is another cancer treated with DC vaccines. In these cases, the patient’s DC cells were

either pulsed with tumor lysate or were pulsed with a mixture of melanoma peptides identified from

animal studies. Some of the melanoma trials produced stronger data than obtained with Provenge. DC

vaccines have also been used to treat glioblastomas (GBMs), a particularly devastating cancer with a

median survival time of less than 2 years (Johnson and O’Neill, 2012). Table-V below shows examples

of experiments with DC vaccines.

Table-V: Example Experiments with DC Vaccines

Cancer Notes Side-Effects Reference

Prostate Cancer Phase-I study. Autologous DCs pulsed with

HLA-A0201-specific prostate-specific

membrane antigen (PSMA) peptides. A

decrease in PSA was observed only in the

group receiving DCs pulsed with peptide P2.

No significant toxicity was

observed for any of the

treatment groups.

Murphy et

al., 1996

Page 50: CANCER VACCINES - Worcester Polytechnic Institute

50

Melanoma Phase-I. Regression of metastases was

observed in 5 of the 16 patients. The vaccinations appeared to

be well tolerated, and did not

generate any visible

autoimmune responses.

Nestle et al.,

1998

Prostate Cancer Phase-I/II. 100% of the patients showed an

immune response against the priming PA-

2024.

The most common side-

effect observed was fever

(14.7% of the patients).

Small et al.,

2000

Prostate Cancer Phase-I. Circulating levels of PSA dropped in 3

of the 13 treated patients. The patients experienced

mild grade-1 and -2 side

effects, such as fever, chills,

myalgia, pain, and fatigue.

Burch et al.,

2000

Melanoma Phase-I. 2 of the 14 patients showed anti-tumor

responses, including regression of metastasis. 1 patient developed vitiligo,

but that skin discoloration

was minor and treatable.

Mackensen

et al., 2000

Melanoma Phase-I. 9 of 12 patients developed CTLs

capable of destroying melanoma cells. The vaccine seemed to be

well tolerated, except 2

patients showed progressive

vitiligo (skin discoloring)

Banchereau

et al., 2001

Glioblastoma Phase-I. 4 out of 7 evaluated patients showed

sustained anti-tumor CTL responses. Median

survival time of the DC group was 455 days

compared to 257 days for the control group.

No serious side effects were

seen. Yu et al.,

2001

Prostate Cancer Phase-II. 2 of the 21 patients showed a 25-50%

drop in PSA levels. One patient dropped PSA

to undetectable levels and resolved his cancer.

Most of the side-effects were

grade-1 and -2, with only 4

of the 21 patients showing

grade-3 or 4 side-effects.

Burch et al.,

2004

Glioblastoma Phase-I. The DCs were primed with tumor

lysates from the same patient. 6 of 10

evaluated patients showed robust T-cell

responses against the tumors.

The vaccines appeared to be

well-tolerated, and no

evidence of autoimmune

disease was seen.

Yu et al.,

2004

Glioblastoma Phase-I. 1 out of 12 patients showed a clinical

response (improved MRI). 6 had measurable

anti-tumor CTL responses, but those did not

translate into clinical responses or prolong

patient survival.

Liau, et al.,

2005

Prostate Cancer Phase-II. 13 of the 18 patients slowed the rate

of increase of their serum PSA levels. None noted. Beinart et al.,

2005

Melanoma Phase-I. 67% of the patients showed CD8+

cells reactive against the priming peptide

G280, and 9 of 9 patients tested had T-cells

that were able to lyse tumor cells in vitro. 3 of

the 9 patients tested showed stable disease, and

2 showed partially stable disease.

None reported. Linette et al.,

2005

Page 51: CANCER VACCINES - Worcester Polytechnic Institute

51

Melanoma Phase-I. 11 of the 15 showed enhancement of

immune responses against the target antigens.

2 out of 9 evaluable patients showed clinical

responses, one showed complete cancer

regression, the other one showed disease

stabilization.

Salcedo et

al., 2006

Prostate Cancer Phase-III. 147 patients treated with Provenge,

and 78 placebo. Found an average 33%

reduction in death for the patients receiving

Provenge versus the placebo (p=0.011).

The most common Provenge-

induced side-effects were all

grade-1 or 2 (mild), lasting

only 1-2 days. They

included: chills, pyrexia

(fever), headache, asthenia

(weakness), dyspnea (labored

breathing), vomiting, and

tremor.

Higano et al.,

2009

Melanoma Review. 626 patients. Improved clinical

responses correlated with the use of peptide

antigens to pulse the DC cells (p=0.03), the use

of adjuvant (p=0.002), and the induction of

antigen-specific T-cells (p=0.0004)

Engell- Noerregaard et al., 2009

Prostate Cancer Phase-III. 341 patients treated with Provenge,

and 171 placebo. Double-blind, placebo

controlled, multicenter trial. Provenge patients

had an average 22% reduction of death, with

survival extending from 21.7 months to 25.8

months.

Adverse effects included

chills, fever, and headache. Kantoff et

al., 2010

Prostate Cancer Summary of 17 Provenge trials. Stable disease

was observed in 54% of the Provenge patients.

High DC doses significantly correlated with

clinical benefit.

Draube et al.,

2011

Melanoma Phase-I. Compared intra-nodal and intra-

dermal vaccinations. All of the intradermal

vaccinated patients showed beneficial DC

migration to the nodes, compared to no

migration for 7 of 24 intra-nodal patients.

Lesterhuis et

al., 2011

Glioblastoma Phase-I. 23 patients with grade-4 GBM were

vaccinated with tumor lysate-pulsed DCs

accompanied by adjuvant. The vaccines

produced a median survival time of 31.4

months, compared to less than 24 in controls.

The vaccines appeared to be

well tolerated. Prins et al.,

2011

Glioblastoma Phase-I. This was the first study to treat

recurrent human malignant gliomas with a

combination of αDC1 cells and adjuvant. Of 19

evaluable patients, 58% had positive immune

responses against the target antigen. 9 patients

had no sign of tumor progression for at least 12

months, and 1 patient showed sustained

complete remission.

The vaccines were well-

tolerated. Okada et al.,

2011

Page 52: CANCER VACCINES - Worcester Polytechnic Institute

52

Melanoma Phase-I. 6 of the 7 patients showed sustained

anti-tumor T-cell responses. 1 patient showed

complete remission, and 2 showed partial

remission. Strong improvements correlated

with with DC cells producing IL-12, and for

patients with strong T-cell responses.

Carreno et

al., 2013

Glioblastoma Pre-Clinical. Pulsing DC cells with glioma

stem cell lysate (these cells often re-seed the

tumor) better forms anti-tumor T-cell

responses than non-stem cell primed DCs.

Ji et al., 2013

Melanoma Phase-I. Used new exome sequencing

technologies to identify neo-antigens present

in the tumor cells, not normal cells. Bio-

informatics was used to identify missense

mutations likely to produce neoantigens. They

selected about 7 neo-antigens per patient,

synthesized them chemically, then charged the

DC cells. T-cell responses were generated

against some, but not all 7 of the neoantigens,

indicating they are not equal in their ability to

induce an immune response.

Carreno et

al., 2015

Glioblastoma Phase-I. Patients were vaccinated with DCs

primed against cytomegalovirus

phosphoprotein-65. Patients also receiving an

adjuvant of tetanus toxoid Td (to generally

boost the immune system) showed a greater

DC migration to the desired location (vascular

draining lymph nodes) than patients without

adjuvant.

Mitchell et

al., 2015

Metastatic

Melanoma Melanoma patients sometimes develop

resistance to immunotherapies via an immune

suppressive tumor microenvironment. Here,

the authors demonstrated in mouse models that

macrophages (MOs) and dendritic cells (DCs)

are suppressed in metastatic melanoma, and

that peptide C36L1 can restore MO and DC

function, including inhibiting metastatic

growth in lungs. The C36L1 treatment

activates MOs, increase the immunogenic DCs,

increase activated cytotoxic T-cells, and reduce

the number of regulatory T-cells in metastatic

lungs. The C36L1 peptide directly binds

receptor CD74 on MOs and DCs, inhibiting

MIF signaling.

Figueiredo et

al., 2018

Colon

Carcionma In this study, the authors investigated the use

of stereotactic body radiation therapy (SBRT)

as a method for facilitating the presentation of

tumor-associated antigens (TAA) to immature

dendritic cells (iDCs). They tested their

method on mouse xenograft models of CT-26

colon carcinoma with 3 groups of mice: 1)

radiation therapy alone (RT), 2) intra-tumor

Choi et al.,

2018

Page 53: CANCER VACCINES - Worcester Polytechnic Institute

53

injection of DCs electroporated with tumor

antigens (DC), or 3) the combination

(RT/iDC). The data showed that the radiation

method achieved the best DC priming and T-

cell activation compared to other priming

methods, and that mouse survival was highest

in the combination treatment group. The

authors conclude that clinical trials are

warranted.

Glioblastoma Phase-III trial of 331 patients with

glioblastoma. The standard therapy for

glioblastoma includes surgery, radiotherapy,

and oral chemotherapy temozolomide. This

study evaluated the addition of an autologous

tumor lysate-pulsed dendritic cell vaccine

(DCVax®-L) to standard therapy for newly

diagnosed glioblastoma patients. After surgery

and chemo-radiotherapy, patients were

randomly assigned to two groups: 1) chemo +

DC vaccine (232 patients), or 2) chemo +

placebo (99 patients). But if the cancer

recurred, the patients were allowed to receive

the DC vaccine. Due to this cross-over design,

nearly 90% of the patients received the DC

vaccine, so patient survival relative to a

placebo could not be determined. However,

median overall survival 34.7 months, with a 3-

year survival of 46.4%.

Only 2.1% (n = 7) of the

patients had a grade 3 or 4

adverse event that was

deemed at least possibly

related to the vaccine.

Overall adverse events with

DC vaccine were comparable

to standard therapy alone.

Liau et al.,

2018

Lewis lung

carcinoma and

breast cancer

cells

In some cases, the tumor microenvironment

can inhibit the activation of the immune system

to fight a tumor, including antigen-pulsed DC

cells. The authors developed a system to

determine whether exosomes (membrane

vesicles) produced by LLC Lewis lung

carcinoma or 4T1 breast cancer cells contribute

to DC immune suppression. They found that

exosomes from these tumors blocked the

differentiation of myeloid precursor cells into

DC cells, and inhibited the migration and

maturation of DCs. The inhibitory response

was partially blocked by treating with anti-PD-

L1 antibody, suggesting this checkpoint

inhibition was important to the inhibition.

Ning et al.,

2018

Diffuse

Instinsic

Pontine Glioma

Phase-Ib clinical trial of 9 patients with diffuse

intrinsic pontine glioma (DIPG), a lethal

brainstem tumor in children. They tested

autologous dendritic cell vaccines (ADCVs)

pulsed with an allogeneic tumor cell-line

lysates in newly diagnosed patients following

radiation therapy. The DCs were prepared

from monocytes obtained by leukapheresis.

The authors found that their procedure boosted

non-specific (KLH) (9/9 patients) and specific

glioma immune anti-tumor responses (8 of 9

The DC vaccine

administration was safe in all

treated patients.

Benitez-

Ribas et al.,

2018

Page 54: CANCER VACCINES - Worcester Polytechnic Institute

54

patients) in PBMCs and in T-lymphocytes

isolated from CSF.

Glioblastoma Glioblastoma (GBM) tumors strongly suppress

the immune system. Checkpoint blockage

vaccines (such as antibodies against PD-1)

might be useful for overcoming the blockade,

but some experiments suggest the checkpoint

approach may not be sufficient. This team

investigated the activation of DC cells as a

supplement to anti-PD-1 therapy in mice.

Their data shows that activating DCs (by

stimulating the TLR3 receptor with poly(I:C)

enhances the PD-1 anti-tumor response and

increases survival in mouse models of

glioblastoma. DC depletion experiments

showed that DCs are required for the anti-

tumor response. The authors conclude that

increasing DC antigen presentation is

important to the anti-tumor response, and that

multi-modal immunotherapy strategies are

important.

Garzon-

Muvdi et al.,

2018

Metastatic

breast cancer The authors investigated the use of

chemotherapy agent Dasatinib as a supplement

for a DC vaccine against metastatic breast

cancer in mice. Their data showed that tumor

volume deceased in the group receiving the

combined treatment, but not in groups

receiving single vaccines. Mouse survival was

longest in the combined treatment group.

Song et al.,

2018

DC Vaccine Future Directions

Overall, while some scientists argue that DC clinical trials have produced somewhat modest

results so far, the data suggests some potential ways for improving efficacy: 1) Adjuvants: Some trials showed improved outcomes combining DCs with adjuvants such as poly(I:C)

(Okada et al., 2011; Ammi et al., 2015), or with IL-12 hormone to boost the immune response (Carreno et

al., 2013), or tetanus toxoid Td (Mitchell et al., 2015). For example, in the latter study, glioblastoma

patients were pretreated at the site of the injection with a recall antigen (in this case, tetanus/diphtheria

(Td) toxoid) with the intention of increasing the efficacy of tumor antigen-specific dendritic cells. Out of

thirteen patients, all of them showed greater accumulation of DCs when given Td than those who were

not pretreated. Three of these patients’ glioblastomas halted progression and had extended survival time.

The overall results of this study concluded that patients given a recall antigen pretreatment had generally

increased survival times compared to patients treated with DCs alone (Mitchell et al., 2015). With further

research, application of adjuvants under different types of circumstances may lead to significantly

improved results.

Page 55: CANCER VACCINES - Worcester Polytechnic Institute

55

2) Tumor Heterogeneity: Some experiments have shown that tumors are not homogenous, but instead

are composed of different types of cells each expressing different neo-antigens. Each type of tumor cell is

genetically distinct from its neighbors, meaning they are different regarding growth rates, metabolic

pathways, and overall aggression, among other characteristics. Perhaps most importantly, certain types of

cells may develop immunity to treatments while others do not. This situation would require several levels

of treatment and decrease the chance that growth will be suppressed or that the tumor will regress on its

own. In these cases, targeting only one antigen would not kill all the tumor cells, so perhaps targeting

multiple antigens would improve efficacy. 3) Which DCs: DC cells are not all alike. Isolating DCs on the basis of surface antigen CD14 selects for

immature cells, but it is not clear whether priming mature or immature DCs is most efficient. Expanding

DCs with hormone Flt3L appears to enrich for a more mature population. Other investigators have had

success using αDC1 cells (Andrews et al., 2008; Okada et al., 2011).

4) Patient Selection: Some patients respond better to DC vaccines than others, but it is not clear

why. Experiments should be done on the tumor micro-environments from various patients to determine if

differences there block immune responses in some patients. For example, the amount of TGF-beta

present in the tumor can strongly affect the immune response (Derynck et al., 2001). 5) Combination Vaccines: To further improve vaccine efficacy, perhaps combinations of cancer

vaccines could be tested, such as combining DC vaccines plus an immune checkpoint vaccine against

CTLA. A clinical study was done to test this mechanism in 16 patients suffering from metastatic

melanoma. The patients were administered the DC vaccine along with a dose of cytotoxic T lymphocyte-

associated antigen 4 (CTLA4)-blocking antibodies. The purpose of adding the antibodies is that they

provide a negative-feedback response to the body’s immune system to promote the activation of T

lymphocytes. The results of this study showed that four patients were entirely tumor-free within two to

four years after the start of the treatment, with no observable relapse. One patient also had total lung

metastasis regression after 4 months, as well as significant (55%) regression of a spinal mass. This

suggests that the combination of both treatments has a better overall result than that of each treatment

individually (Ribas et al., 2009). Other example combination vaccines are Garzon-Muvd et al., 2018 and

Song et al., 2018.

Cited References for Dendritic Cell Vaccines

Ammi R, De Waele J, Willemen Y, Van Brussel I, Schrijvers DM, Lion E, Smits EL (2015) Poly(I:C) as

cancer vaccine adjuvant: knocking on the door of medical breakthroughs. Pharmacol Ther, 2015 Feb;

146: 120-131.

Anagnostou VK, Brahmer JR (2015) Cancer immunotherapy: a future paradigm shift in the treatment of

non-small cell lung cancer. Clinical Cancer Research, 2015 Mar 1; 21(5): 976-984. doi: 10.1158/1078-

0432.

Andrews DM, Maraskovsky E, Smyth MJ (2008) Cancer vaccines for established cancer: how to make

them better? Immunological Reviews, 2008 Apr; 222: 242-255. Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature, 1998 Mar 19;

392(6673): 245-252. Banchereau J, Palucka AK, Dhodapkar M, Burkeholder S, Taquet N, Rolland A, Taquet S, Coquery S,

Wittkowski KM, Bhardwaj N, Pineiro L, Steinman R, Fay J (2001) Immune and clinical responses in

Page 56: CANCER VACCINES - Worcester Polytechnic Institute

56

patients with metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine. Cancer

Research, 2001 Sep 1; 61(17): 6451-6458.

Beinart G, Rini BI, Weinberg V, Small EJ (2005) Antigen-presenting cells 8015 (Provenge) in patients

with androgen-dependent, biochemically relapsed prostate cancer. Clinical Prostate Cancer, 2005 Jun;

4(1): 55-60. Benitez-Ribas D, Cabezón R, Flórez-Grau G, Molero MC, Puerta P, Guillen A, Paco S, Carcaboso AM,

Santa-Maria Lopez V, Cruz O, de Torres C, Salvador N, Juan M, Mora J, La Madrid AM (2018) Immune

Response Generated With the Administration of Autologous Dendritic Cells Pulsed With an Allogenic

Tumoral Cell-Lines Lysate in Patients With Newly Diagnosed Diffuse Intrinsic Pontine Glioma.

Frontiers in Oncology, 2018 Apr 26; 8: 127. doi: 10.3389/fonc.2018.00127. Burch PA, Breen JK, Buckner JC, Gastineau DA, Kaur JA, Laus RL, Padley DJ, Peshwa MV, Pitot HC,

Richardson RL, Smits BJ, Sopapan P, Strang G, Valone FH, Vuk-Pavlović S (2000) Priming tissue-

specific cellular immunity in a phase I trial of autologous dendritic cells for prostate cancer. Clinical

Cancer Research, 2000 Jun; 6(6): 2175-2182.

Burch PA, Croghan GA, Gastineau DA, Jones LA, Kaur JS, Kylstra JW, Richardson RL, Valone FH,

Vuk-Pavlović S (2004) Immunotherapy (APC8015, Provenge) targeting prostatic acid phosphatase can

induce durable remission of metastatic androgen-independent prostate cancer: a Phase 2 trial. Prostate,

2004 Aug 1; 60(3): 197-204. Carreno BM, Becker-Hapak M, Huang A, Chan M, Alyasiry A, Lie WR, Aft RL, Cornelius LA, Trinkaus

KM, Linette GP (2013) IL-12p70-producing patient DC vaccine elicits Tc1-polarized immunity. Journal

of Clinical Investigation, 2013 Aug; 123(8): 3383-3394.

Carreno BM, Magrini V, Becker-Hapak M, Kaabinejadian S, Hundal J, Petti AA, Ly A, Lie WR,

Hildebrand WH, Mardis ER, Linette GP (2015) Cancer immunotherapy. A dendritic cell vaccine

increases the breadth and diversity of melanoma neoantigen-specific T cells. Science, 2015 May 15;

348(6236): 803-808. Choi CW, Jeong MH, Park YS, Son CH, Lee HR, Koh EK (2018) Combination Treatment of Stereotactic

Body Radiation Therapy and Immature Dendritic Cell Vaccination for Augmentation of Local and

Systemic Effects. Cancer Research and Treatment, 2018 Jun 6. doi: 10.4143/crt.2018.186. Dagogo-Jack I, Shaw AT (2017) Tumor heterogeneity and resistance to cancer therapies. Nature Reviews

Clinical Oncology, 2017 Nov 08; 15: 81-94. Dendreon Pharmaceuticals (2017) Provenge Side Effects to Consider. Retrieved June 1, 2018, from

https://www.provenge.com/side-effects Davis ID, Jefford M, Parente P, Cebon J (2003) Rational approaches to human cancer immunotherapy.

Journal of Leukocyte Biology, 2003 Jan; 73(1): 3-29.

Derynck R, Akhurst RJ, Balmain A (2001) TGF-beta signaling in tumor suppression and cancer

progression. Nature Genetics, 2001 Oct; 29(2): 117-129.

Draube A, Klein-González N, Mattheus S, Brillant C, Hellmich M, Engert A, von Bergwelt-Baildon M

(2011) Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and

meta-analysis. PLoS One, 2011 Apr 20; 6(4): e18801.

Page 57: CANCER VACCINES - Worcester Polytechnic Institute

57

Engell-Noerregaard L, Hansen TH, Andersen MH, Thor Straten P, Svane IM (2009) Review of clinical

studies on dendritic cell-based vaccination of patients with malignant melanoma: assessment of

correlation between clinical response and vaccine parameters. Cancer Immunology, Immunotherapy, 2009

Jan; 58(1): 1-14.

Figueiredo CR, Azevedo RA, Mousdell S, Resende-Lara PT, Ireland L, Santos A, Girola N, Cunha

RLOR, Schmid MC, Polonelli L, Travassos LR, Mielgo A (2018) Blockade of MIF-CD74 Signalling on

Macrophages and Dendritic Cells Restores the Antitumour Immune Response Against Metastatic

Melanoma. Frontiers in Immunology, 2018 May 23; 9: 1132. doi: 10.3389/fimmu.2018.01132. Garzon-Muvdi T, Theodros D, Luksik AS, Maxwell R, Kim E, Jackson CM, Belcaid Z, Ganguly S, Tyler

B, Brem H, Pardoll DM, Lim M (2018) Dendritic cell activation enhances anti-PD-1 mediated

immunotherapy against glioblastoma. Oncotarget, 2018 Apr 17; 9(29): 20681-20697. doi:

10.18632/oncotarget.25061.

Higano CS, Schellhammer PF, Small EJ, Burch PA, Nemunaitis J, Yuh L, Provost N, Frohlich MW

(2009) Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active

cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer, 2009 Aug 15; 115(16):

3670-3679. Ji B, Chen Q, Liu B, Wu L, Tian D, Guo Z, Yi W (2013) Glioma stem cell-targeted dendritic cells as a

tumor vaccine against malignant glioma. Yonsei Medical Journal, 2013 Jan 1; 54(1): 92-100. Johnson DR, O'Neill BP (2012) Glioblastoma survival in the United States before and during the

temozolomide era. Journal of Neuro-Oncology, 2012 Apr; 107(2): 359-364. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer

R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF; IMPACT Study Investigators (2010) Sipuleucel-T

immunotherapy for castration-resistant prostate cancer. New England Journal of Medicine, 2010 Jul 29;

363(5): 411-422.

Koski GK, Cohen PA, Roses RE, Xu S, Czerniecki BJ (2008) Re-engineering dendritic cell-based anti-

cancer vaccines. Immunological Reviews, 2008 Apr; 222: 256-276.

Ledford H (2015) Therapeutic Cancer Vaccine Survives Biotech Bust. Nature, 519: 17-18. Lesterhuis WJ, de Vries IJ, Schreibelt G, Lambeck AJ, Aarntzen EH, Jacobs JF, Scharenborg NM, van de

Rakt MW, de Boer AJ, Croockewit S, van Rossum MM, Mus R, Oyen WJ, Boerman OC, Lucas S,

Adema GJ, Punt CJ, Figdor CG (2011) Route of administration modulates the induction of dendritic cell

vaccine-induced antigen-specific T cells in advanced melanoma patients. Clinical Cancer Research, 2011

Sep 1; 17(17): 5725-5735. Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Giovannone AJ, Lin JW, Chute DJ, Mischel

PS, Cloughesy TF, Roth MD (2005) Dendritic cell vaccination in glioblastoma patients induces systemic

and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment.

Clinical Cancer Research, 2005 Aug 1; 11(15): 5515-5525.

Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, Cobbs CS, Heth JA, Salacz M, Taylor S,

D'Andre SD, Iwamoto FM, et al. (2018) First results on survival from a large Phase 3 clinical trial of an

Page 58: CANCER VACCINES - Worcester Polytechnic Institute

58

autologous dendritic cell vaccine in newly diagnosed glioblastoma. Journal of Translational Medicine,

2018 May 29; 16(1): 142. doi: 10.1186/s12967-018-1507-6.

Linette GP, Zhang D, Hodi FS, Jonasch EP, Longerich S, Stowell CP, Webb IJ, Daley H, Soiffer RJ,

Cheung AM, Eapen SG, Fee SV, Rubin KM, Sober AJ, Haluska FG (2005) Immunization using

autologous dendritic cells pulsed with the melanoma-associated antigen gp100-derived G280-9V peptide

elicits CD8+ immunity. Clinical Cancer Research, 2005 Nov 1; 11(21): 7692-7699. Mackensen A, Herbst B, Chen JL, Köhler G, Noppen C, Herr W, Spagnoli GC, Cerundolo V, Lindemann

A (2000) Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in

vitro from CD34(+) hematopoietic progenitor cells. International Journal of Cancer, 2000 May 1; 86(3):

385-392. Mitchell DA, Batich KA, Gunn MD, Huang MN, Sanchez-Perez L, Nair SK, Congdon K5, Reap EA,

Archer GE, Desjardins A, Friedman AH, Friedman HS, Herndon JE 2nd, Coan A, McLendon RE,

Reardon DA, Vredenburgh JJ, Bigner DD, Sampson JH (2015) Tetanus toxoid and CCL3 improve

dendritic cell vaccines in mice and glioblastoma patients. Nature, 2015 Mar 19; 519(7543): 366-369.

Murphy G, Tjoa B, Ragde H, Kenny G, Boynton A (1996) Phase I clinical trial: T-cell therapy for

prostate cancer using autologous dendritic cells pulsed with HLA-A0201-specific peptides from prostate-

specific membrane antigen. The Prostate, 1996 Dec; 29(6): 371-380.

Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D (1998)

Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nature Medicine,

1998 Mar; 4(3): 328-332. Ning Y, Shen K, Wu Q, Sun X, Bai Y, Xie Y, Pan J, Qi C (2018) Tumor exosomes block dendritic cells

maturation to decrease the T cell immune response. Immunology Letters, 2018 Jul; 199: 36-43. doi:

10.1016/j.imlet.2018.05.002. Nobel Prize in Physiology or Medicine (2011)

http://www.nobelprize.org/nobel_prizes/medicine/laureates/2011/# Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, Engh JA, Bartlett DL,

Brown CK, Zeh H, Holtzman MP, Reinhart TA, Whiteside TL, Butterfield LH, Hamilton RL, Potter DM,

Pollack IF, Salazar AM, Lieberman FS (2011) Induction of CD8+ T-cell responses against novel glioma-

associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic

cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with

recurrent malignant glioma. Journal of Clinical Oncology, 2011 Jan 20; 29(3): 330-336.

Palucka K, Banchereau J (2012) Cancer immunotherapy via dendritic cells. Nature Reviews Cancer, 2012

Mar 22; 12(4): 265-277. Prins RM, Soto H, Konkankit V, Odesa SK, Eskin A, Yong WH, Nelson SF, Liau LM (2011) Gene

expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients

vaccinated with dendritic cell immunotherapy. Clinical Cancer Research, 2011 Mar 15; 17(6): 1603-

1615. Ribas A, Comin-Anduix B, Chmielowski B, Jalil J, de la Rocha P, McCannel TA, Ochoa MT, Seja E,

Villanueva A, Oseguera DK, Straatsma BR, Cochran AJ, Glaspy JA, Hui L, Marincola FM, Wang E,

Page 59: CANCER VACCINES - Worcester Polytechnic Institute

59

Economou JS, Gomez-Navarro J (2009) Dendritic cell vaccination combines with CTLA4 blockade in

patients with metastatic melanoma. Clinical Cancer Research, 2009 Oct 01; 15(19): 6267-6276.

Salcedo M, Bercovici N, Taylor R, Vereecken P, Massicard S, Duriau D, Vernel-Pauillac F, Boyer A,

Baron-Bodo V, Mallard E, Bartholeyns J, Goxe B, Latour N, Leroy S, Prigent D, Martiat P, Sales F,

Laporte M, Bruyns C, Romet-Lemonne JL, Abastado JP, Lehmann F, Velu T (2006) Vaccination of

melanoma patients using dendritic cells loaded with an allogeneic tumor cell lysate. Cancer Immunology,

Immunotherapy, 2006 Jul; 55(7): 819-829.

Sallusto F, Lanzavecchia A (2002) The instructive role of dendritic cells on T-cell responses. Arthritis

Research, 4 (Supplement 3): S127–S132.

Schuler G (2010) Dendritic cells in cancer immunotherapy. European Journal of Immunology, 2010 Aug;

40(8): 2123-2130. Small EJ, Fratesi P, Reese DM, Strang G, Laus R, Peshwa MV, Valone FH (2000) Immunotherapy of

hormone-refractory prostate cancer with antigen-loaded dendritic cells. Journal of Clinical Oncology,

2000 Dec 1; 18(23): 3894-3903.

Song N, Guo H, Ren J, Hao S, Wang X (2018) Synergistic anti-tumor effects of dasatinib and dendritic

cell vaccine on metastatic breast cancer in a mouse model. Oncology Letters, 2018 May; 15(5): 6831-

6838. doi: 10.3892/ol.2018.8188.

Steinman RM, Cohn ZA (1973) Identification of a novel cell type in peripheral lymphoid organs of mice.

I. Morphology, quantitation, tissue distribution. Journal of Experimental Medicine, 1973 May 1; 137(5):

1142-1162. Steinman RM, Banchereau J (2007) Taking dendritic cells into medicine. Nature, 2007 Sep 27;

449(7161): 419-426.

Trombetta ES, Mellman I (2005) Cell biology of antigen processing in vitro and in vivo. Annual Review

of Immunology, 2005; 23: 975-1028.

Ueno H, Schmitt N, Klechevsky E, Pedroza-Gonzalez A, Matsui T, Zurawski G, Oh S, Fay J, Pascual V,

Banchereau J, Palucka K (2010) Harnessing human dendritic cell subsets for medicine. Immunological

Reviews, 2010 Mar; 234(1): 199-212. Yu JS, Wheeler CJ, Zeltzer PM, Ying H, Finger DN, Lee PK, Yong WH, Incardona F, Thompson RC,

Riedinger MS, Zhang W, Prins RM, Black KL (2001) Vaccination of malignant glioma patients with

peptide-pulsed dendritic cells elicits systemic cytotoxicity and intracranial T-cell infiltration. Cancer

Research, 2001 Feb 1; 61(3): 842-847.

Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ (2004) Vaccination with tumor lysate-pulsed

dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer

Research, 2004 Jul 15; 64(14): 4973-4979.

Page 60: CANCER VACCINES - Worcester Polytechnic Institute

60

Lit Review Section-3:

TIL Cancer Vaccines

Michaela Hunter

In addition to cancer vaccines that use therapeutic antibodies or dendritic cells (the topics of

previous sections), T-cells are also used as cancer vaccines. T-cells are a type of nucleated white blood

cell that matures in the thymus (thus their name) (although some T-cells also mature in the tonsils)

(Zhang and Bevan, 2011). There are several types of T-cells, including: helper (CD4+), cytotoxic (CD8+),

memory, suppressor, mucosal associated, and gamma delta T-cells.

With respect to cancer vaccines, tumor infiltrating lymphocytes (TILs) are a mixture of T-cells

that locate and infiltrate a tumor to help kill it. TILs isolated from patient tumors include CD4+ (helper T-

cells) and CD8+ cells (cytotoxic T-lymphocytes, CTLs). The CD4+ cells secrete cytokines to activate the

immune system, while the CTLs directly lyse the tumor cell. High levels of TILs in tumors are often

associated with a better clinical outcome (Vanky et al., 1986). T-cell therapy is sometimes referred to as

adoptive cell therapy (ACT) because T-cells are isolated from a patient, expanded in vitro, selected for

particular T-cells targeting a specific antigen, and perfused back (adopted) into the same patient

(reviewed in: Rosenberg and Restifo, 2015; Mayor et al., 2018; Saint-Jean et al., 2018). So, TIL or ACT

therapy is a form of personalized medicine.

So far, TIL therapy can only be performed in a few large medical centers due to the highly

specialized patient care required and the complexity of TIL culture. After isolation of a patient’s TILs,

and before their re-perfusion, the patient is often treated with high-doses of chemotherapy to deplete any

remaining lymphocytes that could block the therapy. And sometimes interleukin-2 (IL-2) injections are

given to increase survival of the perfused T-cells. One advantage of using TILs to fight cancer is that the

in vitro expansion process can sometimes avoid the negative regulation T-cells encounter near the tumor

site by checkpoint inhibition via PD-1, PD-L1, or CTLA-4 (discussed in a later section). So, the ability to

grow and expand T-cells in vitro has been a major advance in the field of cancer therapy.

Examples of TILs and Cancer

Dr. Steven Rosenberg at the National Cancer Institute pioneered the use of TILs to fight cancer,

especially melanomas (for a review, see Rosenberg and Dudley, 2009). His lab helped develop the

procedures for isolating a patient’s TILs and amplifying them in vitro. He also helped develop the

procedure of chemoablation, the use of chemotherapy to deplete a patient’s in vivo lymphocytes that can

suppress TIL function, and then afterwards re-perfusing the therapeutic TILs into the patient. The TIL

studies published so far on melanomas show an impressive clinical response rate of up to 50% with no

side-effects, including a significant proportion of patients with durable complete response (Mayor et al.,

2018). In addition to melanomas, TILs have also been used to fight epithelial and ovarian cancers. Table-

VI below shows some example studies of treating cancer with TILs.

Page 61: CANCER VACCINES - Worcester Polytechnic Institute

61

Table-VI: Example Studies Treating Cancer with TILs

Cancer Type Notes Side-Effects Reference

Metastatic

melanoma 9 of the 41 patients (22%) achieved complete or partial

cancer remissions. Positive outcomes correlated with

high TIL numbers.

Schwartzentruber et

al., 1994 Rosenberg’s team

Metastatic

melanoma Phase-I study, 10 patients. TILs were isolated against

antigens MART1 and gp100 targets. The T-cells

persisted in vivo at least 21 months (using supplement

IL-2) and localized to tumor sites.

No serious

toxicity was

observed.

Yee et al., 2002

Metastatic

melanoma 13 patients. All received chemoablation in advance

and high dose IL2 therapy. 6 of 13 patients (46%)

showed significant tumor regression.

Dudley et al., 2002 Rosenberg’s team

Metastatic

melanoma 2 of the 15 patients showed high levels of circulating

TILs one year after injection, and observable tumor

regression.

Morgan et al., 2006 Rosenberg’s team

Metastatic

Melanoma Patients received pre-treatment chemoablation and/or

total body irradiation, followed by TILs. 49% of the

patients receiving the TILs and no irradiation had an

objective response. Adding 2 Gy irradiation increased

the response to 52%, and adding 12 Gy increased the

response rate to 72%.

Dudley et al., 2008 Rosenberg’s team

Metastatic

melanoma 20 patients received chemoablation and TILs. 50% of

the patients achieved an objective clinical response: 2

complete remissions, and 8 partial remissions.

Manageable

toxicity. Besser et al., 2010

Metastatic

melanoma 93 patients, pre-treated with chemoablation and

radiation. 20 of the 93 patients (22%) achieved

complete tumor regression, and 19 of the 20

regressions remained negative for at least 3 years! The

3 and 5-year survival rates for the 20 remission patients

were 100% and 93%, respectively.

Rosenberg et al.,

2011

Metastatic

melanoma 31 patients. Each received chemoablation and IL-2. 15

of the 31 (48.3%) showed an objective clinical

response.

Radvanyi et al.,

2012

Metastatic

melanoma 19 patients, but only 13 completed the

treatments. Each received chemoablation and IL2. 2 of

13 had complete responses, and 3 had partial responses.

In addition, 4 patients had stable disease.

Pilon-Thomas et al.,

2012

Metastatic

melanoma 69 patients, 35 receiving TILs with no selection and 35

enriched for CD8+ CTLs. 12 patients receiving the

unselected TILs responded to the therapy, while only 7

responded to the CD8+-enriched TILs, so the CD8

enrichment may not be worth the effort.

Dudley et al., 2013 Rosenberg’s team

Page 62: CANCER VACCINES - Worcester Polytechnic Institute

62

Metastatic

melanoma Used whole-exome DNA sequencing of tumor DNA

followed by bioinformatics to identify potential

neoantigens. They synthesized the neo-antigens

synthetically, and tested their recognition by patient

TILs. They identified neoantigens present in about

40% of long-term (5-years) survival patients.

Robbins et al., 2013 Rosenberg’s team

Ovarian cancer Designed a new digital DNA-based assay (termed

QuanTILfy) to count TIL cells and assess their

clonality (percent activity against various antigens).

They demonstrated an association between higher

patient TIL counts and improved patient survival

Robins et al., 2013 Bielas’ team at the

Fred Hutchinson

Metastatic

melanoma Used deep sequencing techniques to determine which

cancer antigens the TIL cells recognized and whether

any TILs expressed inhibitory receptors. Their data

indicated that 6 of 6 analyzed tumors contained TILs

positive for mutated neo-antigens, and all 6 contained

TILs positive for negative immune receptors PD-1,

LAG-3, and TIM-3, indicating that the TILs in their in

vivo state were functionally impaired by the

tumor. Thus, antibody therapy designed against the

negative regulators might improve vaccine

effectiveness.

Gros et al., 2014 Rosenberg’s team

Epithelial

cancer Performed whole exome sequencing on TILs isolated

from epithelial tumors, and showed the TILs

specifically reacted against erbb2-interacting protein

(erbb2ip). When treated with a TIL cell population

where 25% were specific for erbb2ip, the patient

showed a decrease in lesions and disease stabilization.

Tran et al., 2014 Rosenberg’s team

Inflammatory

breast cancer

(IBC)

The authors measured the levels of proteins PD-L1

(checkpoint inhibitor) and CD20 (tumor marker) in 221

biopsies as potential biomarkers of patient

outcomes. The presence of high levels of CD20+ TILs

plus high levels of PD-L1+ TILs was an independent

prognostic factor for patient disease free survival. The

authors suggest pursuing the use of anti-PD-1 or anti-

PD-L1 therapies in these patients.

Arias-Pulido et al.,

2018

Advanced

Melanoma TILs were expanded from excised cutaneous or

subcutaneous metastases and then infused into the

patients who also received subcutaneous IL-2. 9

patients were treated (4 had stage-IIIC melanoma, and

6 had stage-IV melanoma). All but 1 patient had

previously received at least 2 other treatments. The

results showed 1 complete remission, 1 partial

remission, 2 stabilizations, and 6 cancer progressions.

No serious

adverse effects

were reported.

Saint-Jean et al.,

2018

Colorectal

Cancer (CRC) The purpose of the study was to help standardize the

method of evaluating TILs in colorectal cancer (CRC)

clinical trials, as the methods currently differ in each

study. They analyzed 160 patients with Stage II or III

CRC using a new method proposed by the International

TILs Working Group in breast cancer that measured

Iseki et al., 2018

Page 63: CANCER VACCINES - Worcester Polytechnic Institute

63

the area occupied by mononuclear cells over the

stromal area on H&E stained sections. They classified

patients into high-TIL density and low-TIL density

groups. Their results showed that the rates of relapse-

free survival (RFS) and overall survival (OS) in the

high-TILs group were significantly higher than those in

the low-TILs group.

Advanced

Breast Cancer The authors investigated whether the TIL scores taken

from core needle biopsies (CNBs) represent those

taken from resected specimens. They analyzed 220

matched pairs of CNBs and resected specimens,

scoring stromal TILs on slides stained with H&E. The

authors concluded that more than five CNB cores may

accurately predict the TIL score of the entire tumor.

Cha et al., 2018

Metastatic

Urothelial Carcinoma

The authors investigated the prognostic role of TIL

levels on survival in patients with metastatic urothelial

carcinoma (mUC) receiving platinum based

chemotherapy. They analyzed 259 mUC patients, of

which 179 (69%) had intense TILs, and 80 (31%) had

non-intense TILs. The median overall survival was

15.7 months for the intense TIL group versus 6.7

months for the non-intense group (p  < 0.001). The

authors conclude that assaying TIL staining intensity

(numbers) for mUC patients is clinically useful for

patient risk stratification and counseling.

Huang et al., 2018

TIL Problems and Future Directions

The data with TIL therapy shows great promise. In some cases, the teams observed 50% tumor

reduction in about half the patients (Dudley et al., 2008; Besser et al., 2010; Radvanyi et al., 2012; Pilon-

Thomas et al., 2012), and in another study, 22% of the melanoma patients showed complete cancer

remission even 3 years post-treatment (Rosenberg et al., 2011). Some studies noted a direct correlation

between high TIL load and positive patient outcomes (Iseki et al., 2018; Huang et al., 2018), so future

research should focus on methods for further amplifying the cells. And due to poor patient prognosis, the

amplification process needs to be done quickly, so methods to speed amplification are important. In the

case of ovarian tumors, they presented a large variety of neo-antigens, so future experiments should

determine whether amplifying TILs targeting one neo-antigen is sufficient in these cases. In some cases,

the patient’s tumor is found to have very low or no detectable TILs, so future research should also focus

on devising new methods for detecting rare TILs. Immune checkpoint inhibitors can sometimes be a

problem with TILs. One study noted a high expression of negative regulator receptors PD-1, LAG-3, and

TIM-3 on the isolated TILs, so some tumors may be expressing ligands that engage these inhibitory

receptors inactivating the TILs. Future tests should be done with combination treatments of TIL cells

plus an antibody against one of these inhibitory receptors.

Cited TIL References

Arias-Pulido H, Cimino-Mathews A, Chaher N, Qualls C, Joste N, Colpaert C, Marotti JD, Foisey M,

Prossnitz ER, Emens LA, Fiering S (2018) The combined presence of CD20 + B cells and PD-

Page 64: CANCER VACCINES - Worcester Polytechnic Institute

64

L1 + tumor-infiltrating lymphocytes in inflammatory breast cancer is prognostic of improved patient

outcome. Breast Cancer Research and Treatment, 2018 June 1. doi: 10.1007/s10549-018-4834-7.

Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Hershkovitz L, Levy D, Kubi A, Hovav

E, Chermoshniuk N, Shalmon B, Hardan I, Catane R, Markel G, Apter S, Ben-Nun A, Kuchuk I, Shimoni

A, Nagler A, Schachter J (2010) Clinical responses in a phase II study using adoptive transfer of short-

term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clinical Cancer Research,

2010 May 1; 16(9): 2646-2655.

Cha YJ, Ahn SG, Bae SJ, Yoon CI, Seo J, Jung WH, Son EJ, Jeong J (2018) Comparison of tumor-

infiltrating lymphocytes of breast cancer in core needle biopsies and resected specimens: a retrospective

analysis. Breast Cancer Research and Treatment, 2018 Jun 5. doi: 10.1007/s10549-018-4842-7. Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, Topalian SL, Sherry R,

Restifo NP, Hubicki AM, Robinson MR, Raffeld M, Duray P, Seipp CA, Rogers-Freezer L, Morton KE,

Mavroukakis SA, White DE, Rosenberg SA (2002) Cancer regression and autoimmunity in patients after

clonal repopulation with antitumor lymphocytes. Science, 2002 Oct 25; 298(5594): 850-854.

Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE,

Leitman SF, Wunderlich J, Restifo NP, Thomasian A, Downey SG, Smith FO, Klapper J, Morton K,

Laurencot C, White DE, Rosenberg SA (2008) Adoptive cell therapy for patients with metastatic

melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. Journal of

Clinical Oncology, 2008 Nov 10; 26(32): 5233-5239.

Dudley ME, Gross CA, Somerville RP, Hong Y, Schaub NP, Rosati SF, White DE, Nathan D, Restifo

NP, Steinberg SM, Wunderlich JR, Kammula US, Sherry RM, Yang JC, Phan GQ, Hughes MS,

Laurencot CM, Rosenberg SA (2013) Randomized selection design trial evaluating CD8+-enriched

versus unselected tumor-infiltrating lymphocytes for adoptive cell therapy for patients with melanoma.

Journal of Clinical Oncology, 2013 Jun 10; 31(17): 2152-2159.

Gros A, Robbins PF, Yao X, Li YF, Turcotte S, Tran E, Wunderlich JR, Mixon A, Farid S, Dudley ME,

Hanada K, Almeida JR, Darko S, Douek DC, Yang JC, Rosenberg SA (2014) PD-1 identifies the patient-

specific CD8⁺ tumor-reactive repertoire infiltrating human tumors. Journal of Clinical Investigation, 2014

May; 124(5): 2246-2259. Huang HS, Su HY, Li PH, Chiang PH, Huang CH, Chen CH, Hsieh MC (2018) Prognostic impact of

tumor infiltrating lymphocytes on patients with metastatic urothelial carcinoma receiving platinum based

chemotherapy. Scientific Reports, 2018 May 10; 8(1): 7485. doi: 10.1038/s41598-018-25944-1.

Iseki Y, Shibutani M, Maeda K, Nagahara H, Fukuoka T, Matsutani S, Kashiwagi S, Tanaka H, Hirakawa

K, Ohira M (2018) A new method for evaluating tumor-infiltrating lymphocytes (TILs) in colorectal

cancer using hematoxylin and eosin (H-E)-stained tumor sections. PLoS One, 2018 Apr 26; 13(4):

e0192744. doi: 10.1371/journal.pone.0192744. Mayor P, Starbuck K, Zsiros E (2018) Adoptive cell transfer using autologous tumor infiltrating

lymphocytes in gynecologic malignancies. Gynecologic Oncology, 2018 May 23. pii: S0090-

8258(18)30920-X. doi: 10.1016/j.ygyno.2018.05.024. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL,

Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA,

Rosenberg SA (2006) Cancer regression in patients after transfer of genetically engineered lymphocytes.

Science, 2006 Oct 6; 314(5796): 126-129.

Page 65: CANCER VACCINES - Worcester Polytechnic Institute

65

Pilon-Thomas S, Kuhn L, Ellwanger S, Janssen W, Royster E, Marzban S, Kudchadkar R, Zager J,

Gibney G, Sondak VK, Weber J, Mulé JJ, Sarnaik AA (2012) Efficacy of adoptive cell transfer of tumor-

infiltrating lymphocytes after lymphopenia induction for metastatic melanoma. Immunotherapy, 2012

Oct; 35(8): 615-620.

Radvanyi LG, Bernatchez C, Zhang M, Fox PS, Miller P, Chacon J, Wu R, Lizee G, Mahoney S,

Alvarado G, Glass M, Johnson VE, McMannis JD, Shpall E, Prieto V, Papadopoulos N, Kim K, Homsi J,

Bedikian A, Hwu WJ, Patel S, Ross MI, Lee JE, Gershenwald JE, Lucci A, Royal R, Cormier JN, Davies

MA, Mansaray R, Fulbright OJ, Toth C, Ramachandran R, Wardell S, Gonzalez A, Hwu P (2012)

Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-

infiltrating lymphocytes in metastatic melanoma patients. Clinical Cancer Research, 2012 Dec 15;

18(24): 6758-6770. Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E,

Samuels Y, Rosenberg SA (2013a) Mining exomic sequencing data to identify mutated antigens

recognized by adoptively transferred tumor-reactive T cells. Nature Medicine, 2013 Jun; 19(6): 747-752.

Robins HS, Ericson NG, Guenthoer J, O'Briant KC, Tewari M, Drescher CW, Bielas JH (2013) Digital

genomic quantification of tumor-infiltrating lymphocytes. Science Translational Medicine, 2013 Dec 4;

5(214): 214ra169.

Rosenberg SA, Dudley ME (2009) Adoptive Cell Therapy for the Treatment of Patients with Metastatic

Melanoma. Current Opinion in Immunology, 21: 233-240.

Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP,

Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME (2011)

Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer

immunotherapy. Clinical Cancer Research, 2011 Jul 1; 17(13): 4550-4557. Rosenberg SA, Restifo NP (2015) Adoptive cell transfer as personalized immunotherapy for human

cancer. Science, 348: 62-68. Saint-Jean M, Knol AC, Volteau C, Quéreux G, Peuvrel L, Brocard A, Pandolfino MC, Saiagh S, Nguyen

JM, Bedane C, Basset-Seguin N, Khammari A, Dréno B (2018) Adoptive Cell Therapy with Tumor-

Infiltrating Lymphocytes in Advanced Melanoma Patients. Journal of Immunology Research, 2018 Mar

19; 2018:3530148. doi: 10.1155/2018/3530148.

Schwartzentruber DJ, Hom SS, Dadmarz R, White DE, Yannelli JR, Steinberg SM, Rosenberg SA,

Topalian SL (1994) In vitro predictors of therapeutic response in melanoma patients receiving tumor-

infiltrating lymphocytes and interleukin-2. Journal of Clinical Oncology, 1994 Jul; 12(7): 1475-1483. Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K,

Hinrichs CS, Parkhurst MR, Yang JC, Rosenberg SA (2014) Cancer immunotherapy based on mutation-

specific CD4+ T cells in a patient with epithelial cancer. Science, 2014 May 9; 344(6184): 641-645. Vánky F, Klein E, Willems J, Böök K, Ivert T, Péterffy A, Nilsonne U, Kreicbergs A, Aparisi T (1986)

Lysis of autologous tumor cells by blood lymphocytes tested at the time of surgery. Correlation with the

postsurgical clinical course. Cancer Immunology, Immunotherapy, 1986; 21(1): 69-76.

Page 66: CANCER VACCINES - Worcester Polytechnic Institute

66

Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, Greenberg PD (2002) Adoptive T cell

therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma:

in vivo persistence, migration, and antitumor effect of transferred T cells. Proceedings of the National

Academy of Sciences, USA, 2002 Dec 10; 99(25): 16168-16173.

Zhang N, Bevan MJ (2011) CD8(+) T cells: foot soldiers of the immune system. Immunity, 2011 Aug 26;

35(2): 161-168.

Page 67: CANCER VACCINES - Worcester Polytechnic Institute

67

Lit Review Section-4:

CAR Cancer Vaccines

Talal Hamza

In addition to therapeutic antibody vaccines, DC vaccines, and TIL vaccines, are chimeric

antigen receptor vaccines (CARs). CARs, also known as chimeric T-cell receptors, or chimeric

immune-receptors, are a different type of T-cell vaccine than TILs in that the T-cells contain a genetically

engineered T-cell receptor that confers at least two key properties to the T-cells: 1) binding affinity for

the tumor, and 2) signaling properties to activate the T-cells to destroy the tumor (reviewed in: Pule et al.,

2003; Lipowska-Bhalla et al., 2012; Curran et al., 2012; Lim and June, 2017; June et al., 2018; June and

Sadelain, 2018). In this approach, an engineered CAR gene is delivered inside the patient’s own T-cells

in vitro using retroviral vectors, the CAR is expressed on the cell surface, and the engineered T-cells are

delivered back into the same patient. CARs are typically engineered to have a monoclonal antibody-like

affinity for a specific tumor antigen, so they do not rely on formal antigen presentation to recognize the

antigen.

CAR structures have evolved over the years, and are based on the T-cell receptor (TCR) (Figure-

8). The TCR (left panel) consists of extracellular alpha and beta domains associated with CD3

subunits. The T-cell becomes activated when the external TCR domains bind peptides presented by MHC

on the surface of antigen presenting cells or tumor cells. The binding activates signalling via an

intracellular CD3-zeta domain (red). Early CARs (second panel) consisted of antibody-like antigen-

binding domains (antibody variable domains, turquoise), a hydrophobic transmembrane domain, and one

intracellular CD3-zeta signaling domain (red) that becomes activated once the receptor engages the target

antigen. Later generation CARs (third and fourth panels) added additional “co-stimulatory domains”

(such as CD28 or 4-1BB) to help the T-cell divide in vivo. Thus, structurally, CARs combine the

powerful properties of highly specific target antigen recognition (the antibody-like portion), co-

stimulation to increase T-cell survival, and T-cell signaling activation to kill the cancer cell, all combined

in a single engineered receptor molecule (Sadelain et al., 2009).

Figure-8: The Evolution of CAR Structures. Shown are the structures of a typical T-

cell receptor (TCR) (left panel), first-generation (second panel), second-generation (third

panel), and third-generation (fourth panel) CAR cells. Gray denotes the engineered T-

cell. Turquoise represents the extracellular monoclonal antibody variable fragments that

recognize the target antigen, gray represents the hydrophobic transmembrane domain, red

denotes the cytoplasmic CD3-zeta stimulation domain, and turquoise ovals represent the

cytoplasmic co-stimulatory domains such as CD28 and 4-1BB (that help the CAR cells

divide and survive in vivo) (June et al., 2018).

Page 68: CANCER VACCINES - Worcester Polytechnic Institute

68

The foundation of the CAR field was laid in the 1980’s by Israeli immunologist Zelig Eshhar

(reviewed in: Eshhar, 2014), and the technique was further refined by other big-name cancer researchers

such as Steven Rosenberg (National Cancer Institute), Carl June (University of Pennsylvania), and Michel

Sadelain (Memorial Sloan-Kettering Cancer Center). By far, the most successful application of CAR

vaccines to date are those against CD19 for leukemia (reviewed in: Kochenderfer and Rosenberg, 2013;

Jena et al., 2013; June et al., 2018). CARs against CD19 have provided some of the most striking

successes in the entire cancer vaccine field because: 1) CD19 is universally expressed on the surface of all

leukemic B-cells, 2) killing normal B-cells if they happen to express CD19 is not problematic (antibodies

can be provided to the patient passively to compensate for the loss), and 3) CD19 is not expressed outside

the B-cell lineage (so there is little off target killing with the vaccine). Early CAR experiments were

disappointing, and focused on improving CAR design after realizing the injected T-cells quickly die

unless they are co-stimulated in vivo. Table-VII below shows examples of clinical trials using CAR

therapies.

Table-VII: Example Clinical Trial Experiments With CAR Therapies

Target

Antigen Cancer Type Notes Reference

Metastatic

melanoma 15 patients. The CAR cells survived in the peripheral

blood for at least 2 months, and survived for at least

one year in two patients who showed significant tumor

regression.

Morgan et al.,

2006 Rosenberg’s team

α-Folate

Receptor Ovarian cancer Phase-I, 14 patients, but none showed tumor reduction.

PCR analysis showed that the engineered T-cells were

present in the circulation within the first 2 days, but

then quickly declined after one month. Perhaps the T-

cells need a co-stimulator.

Kershaw et al.,

2006 Rosenberg’s team

G-250 Metastatic renal

cell carcinoma 3 patients. Each patient developed liver toxicity so the

treatment had to be discontinued. The patients

continued to show progressive disease. Perhaps the

target antigen is too widespread.

Lamers et al.,

2006

CD20 Lymphoma 3 patients received CARs plus chemoablation to

prevent inhibition of the CARs in vivo. The treatment

was relatively well tolerated. Two patients became

progression-free, with no measurable disease at 12 and

24 months. The third patient initially had a measurable

remission, but relapsed at 12 months.

Till et al., 2008

CD19 Lymphoblastic

leukemia Mouse experiments. Designed a new type of CAR

containing activation domains CD28, CD137

(previously unknown), and/or TCR-zeta. More than

85% of the treated T-cells expressed the engineered

receptor, and the cells survived at least 6 months.

Milone et al.,

2009 Carl June’s team

Page 69: CANCER VACCINES - Worcester Polytechnic Institute

69

CD19 Advanced follicular

lymphoma 1 patient. The first successful clinical treatment of

leukemia using CD19 CARs. Also used a CD28 co-

stimulation receptor. The lymphoma underwent a

dramatic regression, but was this due to the chemo or

the CAR?

Kochenderfer et

al., 2010 Rosenberg’s team

HER-2 (Erbb2)

Colon cancer

metastasized to the

lungs and liver

Tested the vaccine on 1 patient, but he died 5 days after

treatment from a “cytokine storm” (overly elevated

cytokines). The authors speculated that a large number

of perfused CARs localized to the lung cells (with low

but sufficient HER-2) triggering cytokine release. The

study also reminded the researchers that CARs are not

cleared as fast as antibody treatments, so should be

closely monitored.

Morgan et al.,

2010 Rosenberg’s team

CD19 Advanced chronic

lymphocytic

leukemia (CLL)

Breakout year for CARs. CD137 and TCR-zeta co-

stimulators. The CAR cells expanded at least 1000-

fold, migrated to the bone marrow, and continued to

produce functional CARs for at least 6 months, well

beyond the survival of earlier CARs. Each CAR cell

was calculated to destroy about 1,000 cancer cells. One

CLL patient showed complete remission.

Porter et al., 2011 Carl June’s group

CD19 Advanced leukemia 3 CLL patients. Two of the 3 patients showed

complete remission, even 2 years later. Normal B-

cells expressing CD19 were also destroyed causing

grade-3 and 4 B-cell aplasia, but the authors say this

was treatable. These two 2011 studies were later

attributed by Carl June as breaking open the funding

for the entire CAR field.

Kalos et al., 2011 Carl June’s group

CD19 Chronic

lymphocytic

leukemia (CLL)

Also received chemoablation. 8 of the 9 CAR patients

tolerated the treatment well, and 3 of 4 evaluable

patients showed a significant tumor reduction.

Brentjens et al.,

2011 Michel Sadelian’s

team

GD-2 Neuroblastoma Three of 11 patients with active disease achieved

complete remission. Louis et al., 2011

CD19 Relapsed and

refractory acute

lymphoblastic

leukemia (ALL)

2 children with very poor prognosis. The CARs

expanded 1000-fold in vivo, and colonized the bone

marrow and CSF. 1 of the 2 showed complete

remission. The other patient’s tumor lost CD19

expression, so no response. The patients developed

grade-3 and -4 adverse events, including a cytokine

release syndrome, but those events were fully treatable

with cytokine blockade antibodies.

Grupp et al., 2013 Carl June and

Stephan Grupp’s

teams

CD19 Relapsed B-cell

acute

lymphoblastic

leukemia (B-ALL)

5 patients. CD28 and CD3-zeta co-stimulatory

receptors. All 5 patients showed a rapid tumor

eradication, and have no residual disease as assayed

by deep sequencing PCR, although one patient

eventually relapsed. With respect to side-effects, some

patients showed significant cytokine elevations, but

those incidences were treatable with steroid therapy.

Brentjens et al.,

2013 Michel Saedlain’s

team at Sloan

Kettering

Page 70: CANCER VACCINES - Worcester Polytechnic Institute

70

CD19 Chronic

lymphocytic

leukemia (CLL)

None of the patients received prior chemoablation.

PCR showed CD19-CARs in the blood of 8 of the 10

patients. 3 of the 10 treated patients showed

regressions of their previously untreatable

malignancies. One showed complete remission, while

another showed tumor lysis syndrome as his leukemia

regressed.

Kochenderfer et

al., 2013 Steven

Rosenberg’s team

CD19 Refractory B-ALL 16 patients. 88% of the patients responded well enough

to the therapy to later receive a “routine” allogenic

stem cell transplant. They also identified C-reactive

protein (CRP) as a predictor of which patients will

develop severe cytokine release syndrome (sCRS), and

showed they could be treated with corticosteroids and

IL-6-receptor antibodies.

Davila et al.,

2014 Michel Saedlain’s

team

CD19 Relapsed acute

lymphoblastic

leukemia (ALL)

One of the most spectacular successes in all of

cancer vaccine research. 27 of the 30 patients (90%)

showed complete remission, and 67% were event-free

at 6 months. All 30 developed cytokine-release

syndrome (CRS), but it was effectively treated with

anti-interleukin-6 receptor antibody (tocilizumab), and

the patients remained in remission.

Maude et al.,

2014 Dr. Grupp’s team,

Children’s

Hospital,

Philadelphia

CD19 Refractory B-cell

cancers Phase-I. 21 patients. CD28 and TCR-zeta co-

stimulatory receptors. Pre-treatment chemoablation.

The maximum tolerable dose (MTD) was determined

to be 1 x 106 cells per kg, all the side-effects were

reversible at that dose. The most severe side-effects

were a grade-4 cytokine release syndrome observed in

3 of the 21 patients (14%).

Lee et al., 2015

HER-2 Refractory HER-2

Positive Sarcomas

Phase-I/II trial, escalating doses of HER2-CARs with a

CD28 signaling domain. 19 patients. The cell infusions

were well tolerated with no dose-limiting toxicity. The

CARs persisted for at least 6 weeks in 7 of 9 patients

who received a dose of greater than 1 × 10E6 cells per

m2. Of 17 evaluable patients, 4 had stable disease for 12

weeks to 14 months; 3 of these patients had their tumor

removed, with one showing ≥ 90% necrosis.

Ahmed et al.,

2015

CD19 Refractory Multiple

Myeloma 1 patient vaccinated after myeloablative chemotherapy

(melphalan). CAR therapy lead to a complete response,

with no evidence of cancer progression 12 months post-

treatment.

Garfall et al.,

2015

BCMA

(B-cell

maturation

antigen)

Multiple Myeloma First in-humans clinical trial of CARs against

BCMA. 12 patients, various doses. Observed 1 partial

remission at the 3rd dose level, and at the fourth dose

level (9x10E6 CARs/kg body weight) for two patients,

1 showed undetectable cancer for 17 weeks then

relapse, the other patient showed ongoing partial

remission. The highest dose level-4 caused cytokine

release syndrome in both patients, with fever,

hypotension, dyspnea, and cytopenia.

Ali et al., 2016

Page 71: CANCER VACCINES - Worcester Polytechnic Institute

71

CD22

Pre-B-Cell Acute

Lymphoblastic

Leukemia (B-ALL)

Phase-I trial, in 21 children and adults, including 17

who previously treated with CD19-directed

immunotherapy. Complete remission was observed in

11 of 15 (73%) patients receiving more than 1 x 10E6

CD22-CARs per kg body weight. Median remission

duration was 6 months. Relapses were associated with

decreased CD22 expression.

Fry et al., 2018

CEACAM5

Advanced

CEACAM5+

malignancies

The authors tested a first-generation CAR T-cell

therapy. Patients were treated with Fludarabine pre-

conditioning, followed by CEACAM5-CAR T-cells

(various doses), followed by systemic IL2 support. But

no objective clinical responses were observed. T-cell

engraftment showed a rapid decline within 14 days.

Thistlethwaite et

al., 2017

GD2 Diffuse Midline

Gliomas (DMGs) DMGs are aggressive and universally fatal pediatric

brain cancers. The authors showed that patient-derived

DMGs uniformly express high levels of GD2.

Treatment of these cells with GD2-CARs in vitro

showed strong GD2-dependent cytokine generation and

cell killing. The treatment also cleared tumors from 5

patient-derived xenograft mouse models. The treatment

was generally tolerated in mice, but neuro-

inflammation occurred during the acute phase of anti-

tumor activity resulting in hydrocephalus that was

lethal in some animals. They predict that human DMG

patients, given the neuro-anatomical location of the

midline gliomas, will require careful monitoring and

aggressive care management.

Mount et al.,

2018

GD2

ganglioside

Neuroblastoma

The authors generated variant CARs to improve the

stability and the affinity for the target. One variant

(GD2-E101K) showed enhanced antitumor activity in

GD2+ human neuroblastoma xenograft models, but it

also caused lethal CNS toxicity, including brain CAR

T-cell infiltration and proliferation, and neuronal

destruction. The results highlight the challenges

associated with target antigens also expressed on

normal tissues. While GD2-targeted antibody therapies

have shown some success against neuroblastoma, the

fatal neurotoxicity of GD2-CARs suggests additional

strategies are needed for controlling T-cell function in

the brain.

Richman et al.,

2018

CD19

Advanced Lymphoma

Clinical trial with 22 patients receiving a single dose of

CAR-19 T-cells 2 days after a low-dose chemotherapy.

The overall remission rate was 73% with 55% complete

remissions and 18% partial remissions. Remission

patients had a median peak blood CAR+ cell level of

98/μL compared to 15/μL without remission, and the

high CAR levels associated with high serum IL-15

levels (P = .001) and remissions(P < .001).

Kochenderfer et

al., 2017

Page 72: CANCER VACCINES - Worcester Polytechnic Institute

72

CD19 Diffuse Large B Cell Lymphoma

KTE-C19 is an autologous CAR T-cell therapy against

CD19 using CD3-zeta and CD28 co-stimulators. It was

was used to treat 7 patients with refractory DLBCL. 1/7

patients 14%) experienced dose-limiting toxicity of

grade-4 cytokine release syndrome (CRS), 1/7 patients

(14%) showed grade >3 CRS, and 4/7 patients (57%)

showed neurotoxicity. All >grade-3 events resolved

within 1 month. 3/7 patients showed clinical responses

(stabilizations) at 12 months. The regimen appears to

be safe for phase 2 studies.

Locke et al., 2017

An assessment of the number of CAR clinical trials as of January of 2018 (June et al., 2018)

identified 253 CAR trials worldwide (Figure-9). The vast majority of CAR trials are being conducted in

China and the U.S. (left panel). Since China ranks 3rd in the world (behind North America and Europe)

for total clinical trials (right panel), their #1 ranking for CAR trials is quite impressive in this area.

Figure-9: Assessment of CAR Clinical Trials Worldwide. The left panel

shows the number of CAR clinical trials assessed as of January of 2018 (total of

253) compared to the total clinical trials in various countries (right panel) (June

et al., 2018).

CAR Problems and Future Directions

Overall, CAR vaccines have shown some of the most spectacular successes in the entire field of

cancer vaccine research. With so many advances and clinical trial successes, it’s hard not to become

excited. As stated in a review on the topic: “although some scientists are urging caution, it is hard not to

be swept up in this moment. No cell therapy has proliferated in the body as well, endured so well, and

slain cancer, quite like this therapy (Couzin-Frankel, 2013). The overall low number of patients treated

so far likely will improve shortly with the exponential increase in the number of clinical trials. The use of

second and third-generation CARs using a variety of activating and co-stimulating domains appears to

have overcome the earlier problem of CAR cell death in vivo. And pre-treating the patients with

chemotherapy to ablate the endogenous B and T-cells appears to have removed the inhibition against

CAR expansion.

Page 73: CANCER VACCINES - Worcester Polytechnic Institute

73

CRISPR-CAR Combination Therapy One of the most promising and recent innovative uses of CAR cells combines CAR receptor

engineering with the use of CRISPR cas9 to eliminate a host gene. One example showed that using

Lentiviruses and CRISPR greatly increased the efficiency of CAR gene delivery to T-cells. The results

showed CAR expression in human peripheral blood T cells and enhanced effectiveness (Eyquem et al.,

2017), showing better results than typical CAR cells. And in another example, the CRISPR system has

been used to eliminate the gene TRAC which is associated with allo-recognition, and its elimination

might result in CAR cells that could be used universally in any patient (Georgiadis et al., 2018). CD-19

specific CARs were treated with Lentiviruses encoding guide RNAs and Cas9 against TRAC. The

technique produced CAR cells that are far more homogenous than CARs produced using standard

methods (Georgiadis et al., 2018). Furthermore, this combination method produced CAR T-cells with

anti-leukemic effects that are longer lasting than conventionally produced CAR T-cells (Georgiadis et al.,

2018). Ultimately, the combined use of CAR transfections and CRISPR Cas9 transfections is one of the

next steps in CAR T cell therapy.

However, CARs also have problems, including causing patient death in some studies. This is

especially true for CAR treatment of patients with acute leukemia. The following areas are worth

pursuing in the future to make more effective CARs:

1) Side-Effects: The most serious adverse effect observed with CAR trials is patient death (Couzin-

Frankel, 2016; Ledford, 2016). In 2016, 7 trial patients died over about a year due to the CAR therapy

causing fatal brain swelling, and 5 of the 7 patients died in a single clinical trial. Most of the deaths

occurred in adult patients with acute leukemia, and these patients tend to have the worst side-effects due

to rapid expansion of their T-cells (Couzin-Frankel, 2016). The second most serious side-effect is

cytokine release syndrome (CRS), a potentially deadly condition that can cause organ failure (Ledford,

2016). In a large trial of patients with aggressive non-Hodgkin’s lymphoma, about 18% developed CRS

(Ledford, 2016), however this syndrome was often treatable in the patients by blocking the effects of

hormone IL-6 to rapidly reverse the fevers, hypotension, and hypoxia (June et al., 2018).

2) Loss of Target Antigen by the Tumor: The major mode of resistance of some tumors to CAR

therapy is the loss of target antigen by the tumor. This is especially the case for patients with acute

leukemia (June et al., 2018). This underscores the importance of constantly monitoring the patient’s

tumors throughout the entire procedure for target antigen expression, and if a down-regulation occurs,

perhaps the patient could be treated with a CAR targeting a different antigen on the same tumor (if

available). For example, for B-ALL leukemia patients whose tumors no longer expressed CD19, using a

CAR targeting CD22 allowed remissions for at least 6 months (Fry et al., 2018). 3) Failure of the CAR cells to proliferate: The second most common mode of tumor resistance to

CARs is their failure to proliferate in vivo. This is especially the case for patients with chronic cancer

(June et al., 2018). Perhaps these patients would benefit from a combination therapy of CARs plus

checkpoint inhibitor vaccine to activate the immune system. Or alternatively, a CAR using different co-

stimulatory domains would allow CAR proliferation. 4) Correlates of Protection: Some of the studies done with TIL vaccines showed a positive correlation

of a high number of TILs with the best patient prognosis. Is that also true for CARs? Is location

important: Does the location of CARs in the bone marrow affect long-term survival and improve patient

prognosis? Are TILs directed against a patient’s neo-antigens (isolated from the patient’s tumor) better

than CARs directed against a single antigen, such as CD19?

Page 74: CANCER VACCINES - Worcester Polytechnic Institute

74

5) Combination Therapies: TILs isolated from a patient’s tumor can target multiple target antigens

naturally, while artificially engineered CARs target one antigen. Perhaps it would be worth testing both

CARs and TILs in one patient. Or perhaps a combination of a CAR therapy plus a checkpoint inhibitor

treatment could be tested to help block the shutdown of T-cell activity.

6) Personalized Medicine: Neo-antigens are newly expressed proteins or sugars on the surface of tumor

cells that form following DNA mutations in exons (coding DNA). Because neo-antigens are not

expressed during development, they are viewed as foreign once expressed, and make good target antigens

(Delamarre et al., 2015). The use of new rapid DNA sequencing methods allows a patient’s tumor cells

to be sequenced to analyze for neo-antigen formation (Robbins et al., 2013; Rajasagi et al., 2014;

Schumacher and Schreiber, 2015), but the process needs to be fast, as some patients have a very poor

prognosis (Kalos and June, 2013). For example, patients diagnosed with malignant melanoma in stage-IV

can die within weeks.

Cited CAR References

Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, Liu E, Dakhova O, Ashoori A,

Corder A, Gray T, Wu MF, Liu H, Hicks J, Rainusso N, Dotti G, Mei Z, Grilley B, Gee A, Rooney CM,

Brenner MK, Heslop HE, Wels WS, Wang LL, Anderson P, Gottschalk S (2015) Human Epidermal

Growth Factor Receptor 2 (HER2)-Specific Chimeric Antigen Receptor-Modified T Cells for the

Immunotherapy of HER2-Positive Sarcoma. Journal of Clinical Oncology, 2015 May 20; 33(15): 1688-

1696. doi: 10.1200/JCO.2014.58.0225. Ali SA, Shi V, Maric I, Wang M, Stroncek DF, Rose JJ, Brudno JN, Stetler-Stevenson M, Feldman SA,

Hansen BG, Fellowes VS, Hakim FT, Gress RE, Kochenderfer JN (2016) T cells expressing an anti-B-

cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood, 2016 Sep

29; 128(13): 1688-1700. doi: 10.1182/blood-2016-04-711903. Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, Taylor C, Yeh R, Bartido S, Borquez-

Ojeda O, Olszewska M, Bernal Y, Pegram H, Przybylowski M, Hollyman D, Usachenko Y, Pirraglia D,

Hosey J, Santos E, Halton E, Maslak P, Scheinberg D, Jurcic J, Heaney M, Heller G, Frattini M, Sadelain

M (2011) Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients

with relapsed or chemotherapy refractory B-cell leukemias. Blood, 2011 Nov 3; 118(18): 4817-4828. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C,

Olszewska M, Borquez-Ojeda O, Qu J, Wasielewska T, He Q, Bernal Y, Rijo IV, Hedvat C, Kobos R,

Curran K, Steinherz P, Jurcic J, Rosenblat T, Maslak P, Frattini M, Sadelain M (2013) CD19-targeted T

cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic

leukemia. Science Translational Medicine, 2013 Mar 20; 5(177): 177ra38. Brower V (2015) The CAR T-Cell Race. The Scientist, Issue 1 April 2015.

Couzin-Frankel J (2013) The dizzying journey to a new cancer arsenal. Science, 2013 Jun 28; 340(6140):

1514-1518. Couzin-Frankel J (2016) Worries, confusion after cancer trial deaths. Science, 2016 Dec 9; 354(6317):

1211. Curran KJ, Pegram HJ, Brentjens RJ (2012) Chimeric antigen receptors for T cell immunotherapy:

current understanding and future directions. Journal of Gene Medicine, 2012 Jun; 14(6): 405-415.

Page 75: CANCER VACCINES - Worcester Polytechnic Institute

75

Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O,

Olszewska M, Qu J, Wasielewska T, He Q, Fink M, Shinglot H, Youssif M, Satter M, Wang Y, Hosey J,

Quintanilla H, Halton E, Bernal Y, Bouhassira DC, Arcila ME, Gonen M, Roboz GJ, Maslak P, Douer D,

Frattini MG, Giralt S, Sadelain M, Brentjens R (2014) Efficacy and toxicity management of 19-28z CAR

T cell therapy in B cell acute lymphoblastic leukemia. Science Translational Medicine, 2014 Feb 19;

6(224): 224ra25. Delamarre L, Mellman I, Yadav M (2015) Cancer immunotherapy. Neo approaches to cancer vaccines.

Science, 2015 May 15; 348(6236): 760-761. Eshhar Z (2014) From the mouse cage to human therapy: a personal perspective of the emergence of T-

bodies/chimeric antigen receptor T cells. Human Gene Therapy, 2014 Sep; 25(9): 773-778. Eyquem J, Mansilla-Soto J, Giavridis T, van der Stegen SJ, Hamieh M, Cunanan KM, Odak A, Gönen M,

Sadelain M (2017) Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection.

Nature, 2017 Mar 2; 543(7643): 113-117. doi: 10.1038/nature21405. Fry TJ, Shah NN, Orentas RJ, Stetler-Stevenson M, Yuan CM, Ramakrishna S, Wolters P, Martin S,

Delbrook C, Yates B, Shalabi H, Fountaine TJ, Shern JF, Majzner RG, Stroncek DF, Sabatino M, Feng

Y, Dimitrov DS, Zhang L, Nguyen S, Qin H, Dropulic B, Lee DW, Mackall CL (2018) CD22-targeted

CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy.

Nature Medicine, 2018 Jan; 24(1): 20-28. doi: 10.1038/nm.4441. Garfall AL, Maus MV, Hwang WT, Lacey SF, Mahnke YD, Melenhorst JJ, Zheng Z, Vogl DT, Cohen

AD, Weiss BM, Dengel K, Kerr ND, Bagg A, Levine BL, June CH, Stadtmauer EA (2015) Chimeric

Antigen Receptor T Cells against CD19 for Multiple Myeloma. New England Journal of Medicine, 2015

Sep 10; 373(11): 1040-1047. doi: 10.1056/NEJMoa1504542. Georgiadis C, Preece R, Nickolay L, Etuk A, Petrova A, Ladon D, Danyi A, Humphryes-Kirilov N,

Ajetunmobi A, Kim D, Kim JS, Qasim W (2018) Long Terminal Repeat CRISPR-CAR-Coupled

"Universal" T Cells Mediate Potent Anti-leukemic Effects. Molecular Therapy, 2018 May 2; 26(5): 1215-

1227. doi: 10.1016/j.ymthe.2018.02.025. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B,

Wright JF, Milone MC, Levine BL, June CH (2013) Chimeric antigen receptor-modified T cells for acute

lymphoid leukemia. New England Journal of Medicine, 2013 Apr 18; 368(16): 1509-1518.

Jena B, Maiti S, Huls H, Singh H, Lee DA, Champlin RE, Cooper LJ (2013) Chimeric antigen receptor

(CAR)-specific monoclonal antibody to detect CD19-specific T cells in clinical trials. PLoS One, 2013;

8(3): e57838. June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC (2018) CAR T cell immunotherapy for

human cancer. Science, 2018 Mar 23; 359(6382): 1361-1365. doi: 10.1126/science.aar6711.

June CH, Sadelain M (2018) Chimeric Antigen Receptor Therapy. New England Journal of Medicine,

2018 Jul 5; 379(1): 64-73. doi: 10.1056/NEJMra1706169.

Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH (2011) T cells with chimeric

antigen receptors have potent antitumor effects and can establish memory in patients with advanced

leukemia. Science Translational Medicine, 2011 Aug 10; 3(95): 95ra73.

Page 76: CANCER VACCINES - Worcester Polytechnic Institute

76

Kalos M, June CH (2013) Adoptive T cell transfer for cancer immunotherapy in the era of synthetic

biology. Immunity, 2013 Jul 25; 39(1): 49-60.

Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich

JR, Canevari S, Rogers-Freezer L, Chen CC, Yang JC, Rosenberg SA, Hwu P (2006) A phase I study on

adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clinical Cancer Research, 2006

Oct 15; 12(20 Pt 1): 6106-6115. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I, Raffeld

M, Nathan DA, Lanier BJ, Morgan RA, Rosenberg SA (2010) Eradication of B-lineage cells and

regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize

CD19. Blood, 2010 Nov 18; 116(20): 4099-4102. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M, Phan

GQ, Hughes MS, Sherry RM, Yang JC, Kammula US, Devillier L, Carpenter R, Nathan DA, Morgan RA,

Laurencot C, Rosenberg SA (2012) B-cell depletion and remissions of malignancy along with cytokine-

associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood,

2012 Mar 22; 119(12): 2709-2720. Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson

DC, Fowler DH, Hardy NM, Mato AR, Hickstein DD, Gea-Banacloche JC, Pavletic SZ, Sportes C, Maric

I, Feldman SA, Hansen BG, Wilder JS, Blacklock-Schuver B, Jena B, Bishop MR, Gress RE, Rosenberg

SA (2013) Donor-derived CD19-targeted T cells cause regression of malignancy persisting after

allogeneic hematopoietic stem cell transplantation. Blood, 2013 Dec 12; 122(25): 4129-4139. Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM,

Klebanoff CA, Kammula US, Sherman M, Perez A, Yuan CM, Feldman T, Friedberg JW, Roschewski

MJ, Feldman SA, McIntyre L, Toomey MA, Rosenberg SA (2017) Lymphoma Remissions Caused by

Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels.

Journal of Clinical Oncology, 2017 Jun 1; 35(16): 1803-1813. doi: 10.1200/JCO.2016.71.3024. Kochenderfer JN, Rosenberg SA (2013) Treating B-cell cancer with T cells expressing anti-CD19

chimeric antigen receptors. Nature Reviews, Clinical Oncology, 2013 May; 10(5): 267-276. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E

(2006) Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically

retargeted against carbonic anhydrase IX: first clinical experience. Journal of Clinical Oncology, 2006

May 1; 24(13): e20-e22.

Ledford H (2016) Safety concerns blight promising cancer therapy. Nature, 2016 Oct 13; 538(7624): 150-

151. doi: 10.1038/538150a. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R,

Sabatino M, Shah NN, Steinberg SM, Stroncek D, Tschernia N, Yuan C, Zhang H, Zhang L, Rosenberg

SA, Wayne AS, Mackall CL (2015) T-cells expressing CD19 chimeric antigen receptors for acute

lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet, 2015 Feb

7; 385(9967): 517-528. Lim WA, June CH (2017) The Principles of Engineering Immune Cells to Treat Cancer. Cell, 2017 Feb

9; 168(4): 724-740. doi: 10.1016/j.cell.2017.01.016.

Page 77: CANCER VACCINES - Worcester Polytechnic Institute

77

Lipowska-Bhalla G, Gilham DE, Hawkins RE, Rothwell DG (2012) Targeted immunotherapy of cancer

with CAR T cells: achievements and challenges. Cancer Immunology, Immunotherapy, 2012 Jul; 61(7):

953-962. Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, Ghobadi A, Budde LE, Bot A,

Rossi JM, Jiang Y, Xue AX, Elias M, Aycock J, Wiezorek J, Go WY (2017) Phase 1 Results of ZUMA-

1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive

Lymphoma. Molecular Therapy, 2017 Jan 4; 25(1): 285-295. doi: 10.1016/j.ymthe.2016.10.020.

Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E, Liu

H, Wu MF, Gee AP, Mei Z, Rooney CM, Heslop HE, Brenner MK (2011) Antitumor activity and long-

term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood, 2011 Dec 1;

118(23): 6050-6056. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey

SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL,

Grupp SA (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia. New England

Journal of Medicine, 2014 Oct 16; 371(16): 1507-1517. Maude SL, Barrett D, Teachey DT, Grupp SA (2014) Managing cytokine release syndrome associated

with novel T cell-engaging therapies. Cancer Journal, 2014 Mar-Apr; 20(2): 119-122.

Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B,

Danet-Desnoyers G, Campana D, Riley JL, Grupp SA, June CH (2009) Chimeric receptors containing

CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic

efficacy in vivo. Molecular Therapy, 2009 Aug; 17(8): 1453-1464.

Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL,

Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA,

Rosenberg SA (2006) Cancer regression in patients after transfer of genetically engineered lymphocytes.

Science, 2006 Oct 6; 314(5796): 126-129. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA (2010) Case report of a

serious adverse event following the administration of T cells transduced with a chimeric antigen receptor

recognizing ERBB2. Molecular Therapy, 2010 Apr; 18(4): 843-851. Mount CW, Majzner RG, Sundaresh S, Arnold EP, Kadapakkam M, Haile S, Labanieh L, Hulleman E,

Woo PJ, Rietberg SP, Vogel H, Monje M, Mackall CL (2018) Potent antitumor efficacy of anti-GD2

CAR T cells in H3-K27M+ diffuse midline gliomas. Nature Medicine, 2018 May; 24(5): 572-579. doi:

10.1038/s41591-018-0006-x. Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells in

chronic lymphoid leukemia. New England Journal of Medicine, 2011 Aug 25; 365(8): 725-733.

Pule M, Finney H, Lawson A (2003) Artificial T-cell receptors. Cytotherapy, 2003; 5(3): 211-226. Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E, Zhang W, Sougnez C, Cibulskis

K, Sidney J, Stevenson K, Ritz J, Neuberg D, Brusic V, Gabriel S, Lander ES, Getz G, Hacohen N, Wu

CJ (2014) Systematic identification of personal tumor-specific neo-antigens in chronic lymphocytic

leukemia. Blood, 2014 Jul 17; 124(3): 453-462.

Page 78: CANCER VACCINES - Worcester Polytechnic Institute

78

Richman SA, Nunez-Cruz S, Moghimi B, Li LZ, Gershenson ZT, Mourelatos Z, Barrett DM, Grupp SA,

Milone MC (2018) High-Affinity GD2-Specific CAR T Cells Induce Fatal Encephalitis in a Preclinical

Neuroblastoma Model. Cancer Immunology Research, 2018 Jan; 6(1): 36-46. doi: 10.1158/2326-

6066.CIR-17-0211.

Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E,

Samuels Y, Rosenberg SA (2013) Mining exomic sequencing data to identify mutated antigens

recognized by adoptively transferred tumor-reactive T cells. Nature Medicine, 2013 Jun; 19(6): 747-752.

Sadelain M, Brentjens R, Rivière I (2009) The promise and potential pitfalls of chimeric antigen

receptors. Current Opinion in Immunology, 2009 Apr; 21(2): 215-223.

Schumacher TN, Schreiber RD (2015) Neoantigens in cancer immunotherapy. Science, 2015 Apr 3;

348(6230): 69-74. Thistlethwaite FC, Gilham DE, Guest RD, Rothwell DG, Pillai M, Burt DJ, Byatte AJ, Kirillova N, Valle

JW, Sharma SK, Chester KA, Westwood NB, Halford SER, Nabarro S, Wan S, Austin E, Hawkins RE

(2017) The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T

cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer

Immunology, Immunotherapy, 2017 Nov; 66(11): 1425-1436. doi: 10.1007/s00262-017-2034-7. Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, Qian X, James SE, Raubitschek A,

Forman SJ, Gopal AK, Pagel JM, Lindgren CG, Greenberg PD, Riddell SR, Press OW (2008) Adoptive

immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically

modified autologous CD20-specific T cells. Blood, 2008 Sep 15; 112(6): 2261-2271.

Page 79: CANCER VACCINES - Worcester Polytechnic Institute

79

Lit Review Section-5:

Immune Checkpoint Vaccines

Cole Royer

Although some very exciting remissions have been achieved with cancer vaccines, as more

patients have been tested over the years in clinical trials, scientists have come to realize that not all

patients respond to the vaccines. So, recent research has focused on why some tumors are killed and

others are not. One of the most exciting advances in this area is the discovery that T-cells that have

migrated into a patient’s tumor can sometimes become inactivated by immune checkpoint inhibitors

(Hirano et al., 2005; Peggs et al., 2006; Topalian et al., 2012; Sznol and Chen, 2013; Cha et al., 2014;

Herbst et al., 2014). Immune checkpoint inhibitors are receptors, such as PD-1 and CTLA-4, on the

surface of T-cells that bind to inhibitory ligands on other immune cells to inactivate them (reviewed in:

Toplian et al., 2012). Under normal situations, this “checking response” is important for preventing

immune hyper-activation, such as autoimmunity.

In the case of cancer patients, sometimes their tumors present the inhibitory ligands on their

surface, blocking T-cell activation. So, in this case the immune checking response works against the

patient by allowing the tumor to block anti-tumor responses (both native and vaccine) that can kill the

tumor cell. Thus, much vaccine research has focused on using antibodies to block the checkpoint

inhibitors (block the blockers), for example by using antibodies against PD-1 or CTLA-4 to remove the

inhibition against the T-cells that have infiltrated the tumor. This checkpoint inhibitor approach

provides a new and exciting approach for cancer vaccines that is different than other approaches, and is

often used in combination with other therapies. A recent review article on checkpoint vaccines called the

checkpoint therapy approach “arguably one of the most important advances in the history of cancer

treatment” (Ribas and Wolchok, 2018).

PD-1 is inhibitory receptor “Programmed Cell Death-1” located on the surface of activated T-

cells (Freeman et al., 2000; Ribas and Wolchok, 2018). Its ligand is “Programmed Cell Death-Ligand-1

(PD-L1), present on the surface of some tumors and on normal cells exposed to pro-inflammatory

cytokines. When the PD-L1 ligand binds PD-1 receptor, a series of signal transduction events are

activated that decrease T-cell function, such as decreasing T-cell migration and proliferation, restricting

tumor cell killing, and increasing T-cell death (Herbst et al., 2014). Figure-10 below shows how the PD-

1 pathway works to the advantage of tumors. The left panel shows a cancer cell over-expressing PD-L1

protein (blue) on its surface bound to PD-1 receptor (turquoise) on a T-cell. This binding occurs as the T-

cell receptor (dark brown) engages a tumor-specific antigen (yellow) bringing the two cells into close

proximity (McCune, 2018). The right panel shows therapy using antibodies against PD-1 (red) or PD-L1

(yellow) preventing activation of the PD-1 checkpoint pathway, allowing the T-cell to remain active to

kill the tumor cell (McCune, 2018; Abdin et al., 2018).

Page 80: CANCER VACCINES - Worcester Polytechnic Institute

80

Figure-10: Biology of the PD-1 Receptor. The left panel illustrates T-cell

inactivation when the T-cell is brought into close proximity to a tumor cell, the

ligand PD-L1 (blue) on the surface of the tumor cell engages receptor PD-1

(turquoise). The right panel denotes therapy with antibodies against PD-L1

(yellow) or PD-1 (red) binding their respective receptors to prevent activation of

the PD-1 inhibitory signalling pathway. From National Cancer Institute, 2016.

CTLA-4 is the “Cytotoxic T-Lymphocyte-Associated Protein-4” receptor present on the surface

of T-cells. It was discovered in 1987 (Brunet et al., 1987), and when it is bound to its ligands CD80 or

CD86 lowers T-cell activation (Walunas et al., 1994). Activation of the CTLA-4 pathway is often used

by cancer cells to inactivate tumor-infiltrating lymphocytes (TILs) (reviewed in: Peggs et al., 2006; Cha

et al., 2014). CTLA-4 has been demonstrated to have a potent inhibitory role in regulating T-cell

responses. As opposed to inducing cell death like the PD-1 protein, CTLA-4 inhibits T-cell proliferation

and activation upon binding its ligand (usually protein B7) by outcompeting co-stimulatory molecules

like CD28 that are also trying to bind B7 (Figure-11) (Ribas and Wolchok, 2018). Working with a

similar mechanism as PD-1, under normal conditions during immune activation, B7 on an antigen

presenting cell (yellow) engages co-stimulatory receptor CD28 on a T-cell to help activate it. CTLA-4

(purple) is a break to this activation, moving to the cell surface during activation to outcompete CD28 for

binding B7. Unfortunately, this T-cell inhibition pathway is sometimes used by cancer cells presenting

B7 on their surface, leading to a silencing of the T-cell and evasion of the cancer from the immune

system. Checkpoint therapy drugs for CTLA-4 (i.e. Ipilimumab antibody, red) bind CTLA-4 to block

activation of the inhibitory pathway, preventing the tumor from silencing T-cell signaling. This allows

the T-cells to act at their full capacity and attack the tumor (Sharma and Allison, 2015).

Page 81: CANCER VACCINES - Worcester Polytechnic Institute

81

Figure-11: Biology of the CTLA-4 Pathway. During immune activation, an

antigen presenting cell (yellow) binds a T-cell (blue) to induce its differentiation

and activation. In addition to the T-cell receptor engaging the presented antigen

(diagram center), a co-stimulatory pathway involving CD28 and B7 interaction

also occurs to further increase the activation. The CTLA-4 pathway is a break

to this activation, being upregulated in the T-cell to outcompete CD28 for B7.

Cancer cells sometimes over-express B7 to engage the CTLA-4 pathway to

inactivate the T-cell, and therapeutic antibodies against CTLA-4 (red) prevent

this inactivation. From Lee et al., 2012.

The incredible success of checkpoint vaccines is illustrated by the exponential increase in the

number of human clinical trials using these inhibitors (Figure-12). From 2009 to 2017, the number of

clinical trials using checkpoint inhibitors increased from 1 trial (with 136 patients) to 469 trials (with

52,539 patients). The checkpoint treatments are often used in combination with other therapies (lower

row in the diagram), the most frequent being other immune therapies.

Figure-12: Exponential Increase of Clinical Trials

Using Checkpoint Inhibitors. The graph shows the

exponential increase in the patients enrolled in clinical

trials from 2009 to 2017 using checkpoint inhibitors

(upper graph), and the number of clinical trials (middle

row). The lower row denotes the combination drug used

with the checkpoint drug, with the largest cohort being

another immune drug (lower left). Figure is from

Kaiser, 2018.

Page 82: CANCER VACCINES - Worcester Polytechnic Institute

82

Checkpoint Vaccine Examples

Figure-13 below shows the time-line of FDA approval for 6 checkpoint antibodies and one

combination therapy. The green dots show the date of first patient treatment with the drug, and the red

dots indicate the date of FDA approval for using that particular antibody to treat a specific form of cancer.

The first checkpoint antibody used in a patient was Ipilimumab, an antibody against CTLA-4. It was

first used in a patient in June of 2000, and in 2011 was FDA-approved for melanoma patients. The second

checkpoint antibody used was Nivolumab (marketed as Opdivo), an antibody against PD-1. It was first

used on a patient in 2006, and from 2014 to the present has been FDA-approved for treating melanoma

(Topalian et al., 2014), non-small cell lung carcinoma, renal cell carcinoma, Hodgkin’s lymphoma

(Ansell et al., 2015), head and neck cancer, urothelial cancer, high microsatellite instability, and

hepatocellular carcinoma (Gettinger et al., 2015; Rizvi et al., 2015; Tanner, 2015).

Figure-13: Time-Line of FDA Approval for Several Checkpoint

Vaccines. The green dots denote the date of first patient treatment with the

drug, and the red dots indicate the date of FDA approval for using that particular

antibody to treat a specific form of cancer. Figure is from Ribas and Wolchok,

2018.

A recent trend with checkpoint vaccines is their combination with antibodies directed against

tumors, and these studies have produced some spectacular successes. Table-VIII below shows example

studies with checkpoint vaccines.

Table-VIII: Example Studies Using Checkpoint Vaccines

Target Notes Side-Effects Reference

Phase-I study of 39 patients with advanced

metastatic melanoma, colorectal cancer

(CRC), castrate-resistant prostate cancer,

non-small-cell lung cancer (NSCLC), or

renal cell carcinoma (RCC). Anti-PD-1

(MDX-1106) treatment. The results showed

one durable complete response, and two

partial responses. The serum half-life of anti-

PD-1 was 12 to 20 days.

The anti-PD-1 was well

tolerated, with one serious

adverse event of

inflammatory colitis.

Brahmer et al.,

2010

Page 83: CANCER VACCINES - Worcester Polytechnic Institute

83

PD-1

Phase-I multi-center Phase-I trial of anti-PD-

L1 therapy, 207 patients with non-small-cell

lung cancer, melanoma, colorectal cancer,

renal-cell cancer, ovarian cancer, pancreatic

cancer, gastric cancer, and breast cancer.

Among patients with a response that could

be evaluated, an objective response (a

complete or partial response) was observed

in 9 of 52 patients with melanoma, 2 of 17

with renal-cell cancer, 5 of 49 with non-

small-cell lung cancer, and 1 of 17 with

ovarian cancer.

Grade-3 or 4 toxic effects

related to treatment occurred

in 9% of patients.

Bramer et al.,

2012

International Phase-II study, anti-PD-1

monoclonal antibody, in 36 patients with

diffuse large B-cell lymphoma

(DLBCL). Among the 35 patients with

measurable disease after stem cell transplant,

the overall response rate after antibody

treatment was 51%.

Toxicity was mild.

Armand et al.,

2013

135 patients with advanced melanoma, anti-

PD-1 antibody treatment in patients

previously receiving CTLA-4 antibody and

those who had not. The confirmed response

rate across all dose cohorts was 38% The

response rate did not differ significantly

between patients who had received prior

anti-CTLA-4 treatment and those who had

not.

Common adverse events

attributed to treatment were

grade-1 or 2, including

fatigue, rash, pruritus, and

diarrhea.

Hamid et al.,

2013

International, multi-center Phase-I trial of

173 patients with advanced melanoma

previously treated with anti-CTLA-4

antibody. The overall response rate was

26%.

The treatment was well

tolerated. There were no

drug-related deaths. The most

common drug-related adverse

events in the 10 mg/kg group

were fatigue (37%), pruritus

(19%), and rash (18%).

Grade-3 fatigue was reported

in five (3%) patients.

Robert et al.,

2014

The treated 107 melanoma patients showed a

mean overall survival of 16.8 months, with

1- year and 2-year survival rates of 62% and

43%, respectively.

The antibody safety was

“acceptable”, with toxicity

rates similar to previous

reports.

Topalian et al.,

2014

Phase-III controlled study, Nivolumab anti-

PD-1 therapy versus dacarbazine

chemotherapy, 418 patients with metastatic

melanoma. At 1 year, the overall rate of

survival was 72.9% in the antibody group

compared to 42.1% in the chemotherapy

group.

Common adverse events

associated with the antibody

treatment included fatigue,

pruritus, and nausea, with

grade-3 or 4 events in 11.7%

of the patients treated with

antibody and 17.6% treated

with chemotherapy.

Robert et al.,

2015

Page 84: CANCER VACCINES - Worcester Polytechnic Institute

84

23 patients with relapsed or refractory

Hodgkin's lymphoma received Nivolumab

anti-PD-1 treatment. A high percent of the

patients had received previous treatments

with stem-cell transplantation or

Brentuximab vedotin (antibody-drug

conjugate targeting CD30). An objective

response was reported in 87% of the

patients, including 17% with a complete

response and 70% with a partial response.

Drug-related adverse events

of any grade occurred in 78%

of the patients, grade-3 events

in 22%. and of grade 3

occurred in 78% and 22% of

patients, respectively.

Discontinuation of the study

due to the drug occurred in 2

patients.

Ansell et al.,

2015

Phase-II study of 22 patients with non-small

cell lung cancer (NSCLC). the drug

produced “durable immune responses and

encouraging survival rates

14% of the patients showed

grade-3 or -4 treatment-

related adverse events.

Gettinger et al.,

2015

Non-small cell lung cancer patients.

Analyzed non-synonymous mutations to

show that higher neo-antigen formation

(more targets) correlated with better clinical

benefits and progression-free patient

survivals.

Rizvi et al.,

2015

600 patients with non-small cell lung cancer.

The median survival rate increased from 9

months (chemotherapy alone) to 12 months

(for the immunotherapy group), and the

tumors shrank in 12% of the chemotherapy

patients versus 20% of the immunotherapy

patients.

Tanner, 2015

Phase-I trial for 655 patients with advanced

metastatic melanoma using humanized anti-

PD-1 monoclonal antibody Pembrolizumab,

performed in academic medical centers in

Australia, Canada, France, and the United

States. An objective response was reported

in 194 of 581 patients (33%). 44% (90/205)

had response duration of at least 1 year.

Ribas et al.,

2016

Multicenter Phase-II trial of 315 patients

with metastatic urothelial carcinoma that

have failed standard platinum chemotherapy

using humanized antibody against PD-

L1. The antibody-treated group showed an

overall response rate of 15%, compared to a

historical response rate of 10%.

Grade 3-4 treatment-related

adverse events occurred in 50

(16%) of 310 treated patients.

Fatigue was the most

common (5 patients, 2%). No

treatment-related deaths

occurred during the study.

Rosenberg et

al., 2016

112 melanoma patients treated with anti-PD-

1 therapy. Identified oral and gut

microbiome differences in patient responders

versus non-responders. Responder gut

microbiomes showed greater microbial

diversity, higher abundance of

Ruminococcaceae, and enrichment of

anabolic pathways. Mice receiving patient

Gopalakrishnan

et al., 2018

Page 85: CANCER VACCINES - Worcester Polytechnic Institute

85

responder fecal transplants showed better

vaccine responses.

Determined that a primary mode of epithelial

cancer patient resistance to anti-PD-1

therapy is the gut microbiome

composition. Antibiotics tended to lower the

clinical benefit the therapy, while fecal

microbiota transplantation (FMT) from

responder patients into mice improved the

treatment. Responders tended to have high

levels of Akkermansia muciniphila, and

supplementation with this microbe tended to

improve outcomes in mouse models.

Routy et al.,

2018

The team developed mouse models of

colorectal cancer with increased levels of

TGFβ in the tumor microenvironment to

mimic patients with T-cell exclusion and

resistance to anti-PD-1 therapy. The mice

showed limited response to anti-PD-1

therapy. Blocking the TGFβ signaling

pathway unleashed a potent and enduring

cytoxoxic T-cell response against the tumors

and improved the anti-PD-1 response. Thus,

TGFβ in the tumor microenvironment is a

primary mechanism of tumor immune

evasion.

Tauriello et al.,

2018

Performed a risk analysis for anti-PD-1

Nivolumab for hepatotoxicity. Analyzed all

Phase-I through Phase-III trials through

December 2016 for elevated levels of liver

enzymes in the blood: aspartate

aminotransferase (AST) and alanine

aminotransferase (ALT). Overall

hepatotoxicity was only 5.4%, and high-

grade toxicity was 1.6%.

Zarrabi and Wu,

2018

CTLA-4

9 patients with various cancers. CTLA-4

antibody treatment caused extensive tumor

necrosis in three of three (100%) metastatic

melanoma patients, and the reduction or

stabilization of cancer in two of two (100%)

metastatic ovarian carcinoma patients, but

caused no reduction in 4 of 4 metastatic

melanoma patients previously immunized

with defined melanoma antigens.

No serious toxicities directly

attributable to the antibody

therapy were observed.

Hodi et al.,

2003

Phase-I trial. 39 patients with solid

malignancies (34 melanoma, 4 renal cell,

and 1 colon, n = 1). The maximum tolerated

dose (MTD) was determined to be 15

mg/kg. 2 patients experienced complete

responses, and two experienced partial

responses.

Dose-limiting toxicities and

autoimmune phenomena

included diarrhea, dermatitis,

vitiligo, pan hypo-pituitarism

and hyper-thyroidism.

Ribas et al.,

2005

Page 86: CANCER VACCINES - Worcester Polytechnic Institute

86

CTLA-4

Metastatic melanoma patients. The median

overall survival of patients receiving the

CTLA-4 antibody + gp100 peptide vaccine

was 10 months compared to 6.4 months for

those receiving only the gp100.

The side effects were severe

in only 10-15% of the

patients receiving both

treatments, and in only 3% of

the patients receiving

gp100. Most were

manageable. 2.1% of the

patients died from the study,

7 related to immune adverse

events.

Hodi et al.,

2010

The article discusses the

different kinds of immune-

related adverse events

associated with CTLA-4

treatment, and the necessary

treatments to manage them.

They conclude that one of the

most important aspects of

side-effect treatment is early

recognition.

Weber et al.,

2012

11 melanoma patients. Performed whole

exome sequencing was performed to

correlate which neo-antigens correlate with

best improvements using CTLA-4 antibody

treatments.

Snyder et al.,

2014

Tested advanced melanoma patients with

CTLA-4 antibody if they did not respond to

PD-1 antibody. 31.7% showed positive

responses compared to 10.6% with

chemotherapy.

Noted grade-3 to 4 (serious)

adverse events in 5% of

antibody-treated patients

versus 9% for chemotherapy.

There were no treatment-

related deaths.

Weber et al.,

2015

Metastatic osteosarcoma. The tumor burden

decreased, and the survival rate increased.

Lussier et al.,

2015

A pooled analysis of overall survival data

from 1,861 patients from 10 prospective and

two retrospective studies of ipilimumab,

including 2 phase-III trials. Patients were

previously treated (n = 1,257) or treatment

naive (n = 604), and the majority of patients

received ipilimumab 3 mg/kg (n = 965) or 10

mg/kg (n = 706). Of the 1,861 patients, the

median overall survival was 11.4 months,

including 254 with at least 3 years of

survival.

Schadendorf et

al., 2015

A review of the data of 4 Phase-I and II trials

at 2 sites for 143 patients with advanced

melanoma. The median overall survival was

13 months, with a 5 year survival rate of

20%, and a 12.5 year survival rate 16%.

Eroglu et al.,

2015

Page 87: CANCER VACCINES - Worcester Polytechnic Institute

87

Analyzed 536 metastatic cancer patients

treated with anti-PD-1, PD-L1, or CTLA-4

immunotherapies for liver related side-

effects. Only 19 patients (3.5%) needed

referral to the liver unit for grade ≥3

hepatitis. No patients developed hepatic

failure. 6 patients improved spontaneously;

the remainder received various doses of oral

corticosteroids. Liver biopsy was helpful for

the diagnosis and evaluation of the severity

of liver injury. The severity of liver injury

was helpful for tailoring patient

management, which does not require

systemic corticosteroid administration.

De Martin et al.,

2018

Combination

Treatments

PD-1 + CTLA-4. Melanoma patients. 53%

of the combination patients showed a tumor

reduction of 80% or more, compared to only

20% for single antibody treatments.

Grade-3 and 4 side-effects

occurred in 53% of the

combination patients

compared to 18% in the

single antibody patients. The

side-effects were generally

reversible.

Wolchok et al.

2013

PD-1 + CTLA-4. Various cancers. Inverse

treatments, used the opposite antibody

against patients showing resistance to the

first antibody.

Found that in most cases, the

second antibody was well

received.

Weber et al.,

2013

PD-1 + CTLA-4. Studied metastatic

osteosarcoma in mice. The combination

treatment prevented tumor escape and

allowed complete control of the cancer.

Lussier et al.,

2015

Phase-III trial of 1096 patients with

advanced clear-cell renal-cell carcinoma

with antibodies to PD-1 + CTLA-4. At 25.2

months, the 18-month survival rate was 75%

for the combination versus 60% for

treatment with a tyrosine kinase inhibitor.

Treatment-related adverse

events occurred in 509 of 547

patients (93%) in the

combination group versus

521 of 535 patients (97%) in

the kinase inhibitor group.

Treatment-related adverse

events causing termination

from the trial occurred in

22% and 12% of the patients,

respectively.

Motzer et al.,

2018

Cancer cells sometimes express TGF-β

which activates regulatory T-cells to inhibit

CD8+ cytotoxic T-cells. The authors

developed a bi-specific antibody, with one

arm recognizing CTLA-4 or PD-1, and the

other arm representing the ectodomain of the

TGFβ receptor II (to bind and sequester

TGFβ). The bi-specific therapy was more

effective at inducing tumor regression (and

reducing regulatory T-cells) than the mono-

specific antibody.

Ravi et al., 2018

Page 88: CANCER VACCINES - Worcester Polytechnic Institute

88

Checkpoint Vaccine Problems and Future Directions

Some of the data using checkpoint inhibitor vaccines is striking, including some complete

remissions from very difficult cancers. The data also shows that using a combination of antibodies (for

example against PD-1 and CTLA-4, or combining PD-1 with a bi-specific antibody) may produce better

data than using either antibody alone. But as groundbreaking and promising as immune checkpoint

vaccines have been, several factors are drawbacks to their use. Blocking the immune blockers can

sometimes activate the immune system too far, producing autoimmunity or inflammation (Melero et al.,

2007; Frankel, 2017), so inflammation should be constantly monitored during the treatments. Other

studies showed the treatments caused grade-3 and -4 (serious) side-effects, some even resulting in death

(Eggermont et al., 2016). Although most of the side-effects appeared to be transient and treatable. The

side-effects were especially treatable if they were detected early, so these events should be constantly

monitored throughout the entire treatment. In other cases, the patient simply did not respond to the

treatment. Even in studies with the best objective response rates, roughly 30% of patients would see no

response. Finally, as with many aspects of our healthcare system, the cost of these drugs can put them out

of reach for many patients, with one course costing up to $150,000 (McCune, 2018). These problems

should be addressed with future experiments.

High Incidence of Complications/Side-Effects

In nearly all the clinical trials examined in this section of the Literature Review, regardless of

drug, dosage, or disease, a large percentage of patients suffered severe (grade 3 or 4) side-effects caused

by the treatment. One study saw nearly half of the patients with side-effects, with 1 in 5 patients being

grade-3 or 4, while another study saw 68% with adverse effects, with 1 patient in 10 being serious

(Topalian et al., 2012; Weber et al., 2015). Another study noted that while most of the side-effects were

manageable, 2.1% of the patients died from the study, 7 related to immune adverse events (Hodi et al.,

2010).

When combining the individual drugs Ipilimumab and Nivolumab into one therapy, the rate of

grade-3 and 4 adverse effects was 53% compared to 18% in the single antibody patients (Wolchok et al.,

2013). Another combination trial observed adverse events in 93% of 547 patients, with those events

causing termination from the trial in 22% of the patients (Motzer et al., 2018). While these adverse

incidences are troubling, they are not out of the norm for cancer therapies. Indeed, some of the trials

reported less frequent occurrence of severe adverse effects than standard chemotherapy (Abdin et al.,

2018). And in most cases, the side-effects were transient and treatable, with a prognosis far better than

the terminal cancer being treated. However, any treatment where a large portion of patients have high

grade complications is not ideal, and this keeps the checkpoint vaccines from being a first-line treatment.

Autoimmune Disorders

Most of the adverse side-effects described above were autoimmune related. The most commonly

observed were thyroid disease, type-1 diabetes, colitis, and liver damage. Checkpoint inhibitors dial up

the immune system beyond physiologically normal levels to overcome the mechanisms that tumors

deploy to hide from the immune cells, so it is natural to see how autoimmune disorders could rise from

this strengthened immune system. The checkpoint inhibitor antibodies can act on normal cells as well the

cancer, allowing the immune system to overcome an important block for the body (Frankel, 2017). Liver

damage (hepatitis) was a particularly well reported side effect, with one study seeing 3.5% of all

checkpoint patients with hepatitis severe enough to require hospitalization (De Martin et al.,

2018). Another study with a 20% rate of grade-3 or 4 complications showed 10.3% with liver toxicity

(Zarrabi and Wu, 2018). One of the more serious autoimmune diseases seen was autoimmune

myocarditis, an immune attack on the heart which can be fatal (Frankel, 2017). Fortunately, this was rare

and most of these autoimmune side-effects were treatable and preferable to progression of

Page 89: CANCER VACCINES - Worcester Polytechnic Institute

89

cancer. However, their burden and potential fatality must be taken into consideration before checkpoint-

based treatment can be initiated (Zarrabi and Wu, 2018).

Low Response Rate

The clinical trials examined here show that even in the best scenarios, up to one in three patients

had no response to the checkpoint therapy. And some types of cancer simply do not appear to respond to

checkpoint inhibitors (Ravi et al., 2018). New research is focusing on how the specific genetic makeup

and personal microenvironment of the patient’s tumor and gut can determine how effective the checkpoint

therapy might be, hopefully identifying a variety of molecular and microbial biomarkers that can be used

to personalize therapy and determine which patients will best respond.

A recent fascinating finding relates to the hormone transforming growth factor-beta (TGF-β) and

its potential role in determining the receptibility of a tumor to a checkpoint therapy. TGF-β is a multi-

functional cytokine with a critical role in regulating the adaptive immune system. When secreted into the

bloodstream during an immune response, TGF-β restricts the differentiation of helper T-cells, stops the

development of memory T-cells, and induces the production of regulatory T-cells during an immune

response. Like PD-1 and CTLA-4, TGF-β protects healthy somatic cells from an overactive immune

system and prevents an autoimmune response. However, many tumors also over-express TGF-β to evade

the immune system. In tumors that do this, checkpoint therapy targeting PD-1 has not been effective;

although the PD-1/PD-L1 system was inactivated by the therapy, the related cytokine response was still

keeping the immune system in check in the tumor microenvironment. The best results for these tumors

came when an antibody targeting TGF-β was used in conjunction with a checkpoint inhibitor to block

both mechanisms of immune system evasion (Ravi et al., 2018). Another study supporting these results

showed that a tumor microenvironment with high levels of TGF-β had almost no response to checkpoint

therapy, but showed a potent response after inhibition of TGF-β (Tauriello et al., 2018). These results

point to the potential use of TGF-β levels as a biomarker for identifying which specific patients have a

cancer microenvironment that would respond to checkpoint therapy alone and those that would require a

combination treatment.

Other recent work has shown that the bacterial composition of the patient’s gut microbiome can

modulate the effectiveness of checkpoint inhibitors. Bacterial mediated interactions with the immune

system are essential for its normal function, and by extension optimal checkpoint inhibitor therapy

outcomes (Jobin, 2018). The gut provides an environment that houses many immune cells. Beneficial gut

bacteria can cause these immune cells to secrete antibodies. Patients who responded to checkpoint

therapy were shown to have functional differences in the types of bacteria that predominated their gut

relative to patients that did not respond (Gopalakrishnan et al., 2018). One study found that patients who

responded to Nivolumab had an abundance of bacteria of the genus Faecalibacterium, while non-

responders had a much lower abundance of this microbe. This team also showed that targeted microbial

enrichment of non-responding patients with the Faecalibacterium correlated with a greater response to

PD-1 therapy and an increase of T-cells in the tumor (Jobin, 2018). Another study showed that resistance

to checkpoint therapy can be attributed to an abnormal gut microbiome. When antibiotics were

prescribed to return the gut microbiome to its optimal state, the effectiveness of checkpoint inhibitors

increased (Routy et al., 2018). The same study correlated the abundance of the species Akkermansia

muciniphila to checkpoint efficacy, as oral supplementation of this microbe increased therapy results

(Routy et al., 2018). These studies point to the composition of a patient’s gut fauna as a promising

indicator of whether they will respond to checkpoint therapy, and also provide hope that those who do not

have an optimal gut microbiome can be given supplemental treatment that will increase their chances of a

positive response.

A third variable that has received increased attention for its role in improving checkpoint therapy

effectiveness is the human leukocyte antigen (HLA) class of molecules. The HLAs are a set of cellular

surface proteins that immune cells use to distinguish the body’s cells from foreign cells. Every person has

Page 90: CANCER VACCINES - Worcester Polytechnic Institute

90

a slightly different set of HLAs that their immune system uses as markers, and the exact makeup of these

proteins is determined by the genotype of the HLA gene complex (American Cancer Society, 2017). One

study demonstrated that melanoma patients with increased heterozygosity in their HLA genes had

increased survival rates compared to patients with a high level of HLA homozygosity (Chowell et al.,

2018). A meta-analysis of the outcomes of over 1,500 cancer patients treated with checkpoint therapy

supported these conclusions, finding that better outcomes often occurred in patients with HLA subtype

heterozygosity. These results make sense because checkpoint therapy depends on the ability of the

immune system to recognize a tumor as an invading cell, and this is easier if the somatic cells have a

more complex and varied identification system (from heterozygosity of the HLA genes). It is easier for a

tumor to avoid detection when HLA is homozygous because the identification proteins on somatic cells

will be less complex and easier to mimic. The same study also showed that a specific subtype of HLA

(HLA-B44) gave the best outcomes for the checkpoint patients, and the HLA-B62 subtype gave a poor

outcome. It is not well understood why the subtypes affected checkpoint therapy this way, but this work

showed that a patient’s HLA genetic makeup might be used as a biomarker to predict effectiveness of the

checkpoint therapy (Chowell et al., 2018).

As this last section has shown, numerous factors beyond the type and stage of the cancer affect

the success of checkpoint therapy. Individual differences in the tumor microenvironment (TGF-β levels),

the gut microbiome, and patient HLA immunogenetics can explain why two people with the same disease

respond so differently to therapy. These individualized determinants have led to the rise of personalized

therapy using these individual differences as biomarkers to indicate whether a patient will respond well to

the checkpoint inhibitor therapy. However, the use of biomarkers is not perfect. Ledford 2018 showed

that biomarker tests can be inconclusive or give false negatives. Nevertheless, individual differences in

patients and their tumors have been clearly demonstrated to determine the effectiveness of

immunotherapy, even though such experiments remain in their infancy. More work to solidify the

identification of biomarkers and the patients best fit for checkpoint therapy is needed for providing more

positive data for the FDA approval of more immune treatments (Leford, 2018).

Recent Failed Clinical Trial

The checkpoint vaccine field also recently suffered a setback with the failure of one of its Phase-

III clinical trials (Garber, 2018). The Phase-III trial was being conducted by the biotech company Incyte

using a combination treatment of Opdivo (PD-1 antibody) and Epacadostat. Epacadostat blocks the

enzyme indoleamine 2,3-dioxygenbase (IDO) which is thought to become activated in T-cells that have

been treated with a checkpoint inhibitor creating a “negative feedback loop” to put a brake on the

activated T-cells. But this feedback loop works against the cancer treatment, so scientists thought that

inhibiting IDO might allow a more prolonged activation of the T-cells. In earlier Phase-I and Phase-II

trials, the combination treatment worked well, but in the recent Phase-III trial the combination was no

better than Opdivo alone. So, maybe the IDO enzyme doesn’t really block the activated T-cells as

scientists believed? Or maybe the wrong patients were treated? Unfortunately, the Incyte failure caused

three other companies to cancel, suspend, or downsize their Phase-III trials with Epacadostat (Garber,

2018).

Cited References for Immune Checkpoint Vaccines

Abdin SM, Zaher DM, Arafa EA, Omar HA (2018) Tackling Cancer Resistance by Immunotherapy:

Updated Clinical Impact and Safety of PD-1/PD-L1 Inhibitors. Cancers (Basel), 2018 Jan 25; 10(2): E32.

doi: 10.3390/cancers10020032.

American Cancer Society Staff (2017). Immune Checkpoint Inhibitors to Treat Cancer. American Cancer

Society, 2017. www.cancer.org/treatment/treatments-and-side-effects/ treatment-types/

immunotherapy/immune-checkpoint-inhibitors.html

Page 91: CANCER VACCINES - Worcester Polytechnic Institute

91

Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM,

Cattry D, Freeman GJ, Rodig SJ, Chapuy B, Ligon AH, Zhu L, Grosso JF, Kim SY, Timmerman JM,

Shipp MA, Armand P (2015) PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's

lymphoma. New England Journal of Medicine, 2015 Jan 22; 372(4): 311-319. Armand P, Nagler A, Weller EA, Devine SM, Avigan DE, Chen YB, Kaminski MS, Holland HK, Winter

JN, Mason JR, Fay JW, Rizzieri DA, Hosing CM, Ball ED, Uberti JP, Lazarus HM, Mapara MY,

Gregory SA, Timmerman JM, Andorsky D, Or R, Waller EK, Rotem-Yehudar R, Gordon LI (2013)

Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous

hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international

phase II trial. Journal of Clinical Oncology, 2013 Nov 20; 31(33): 4199-4206. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay

TM, McMiller TL, Gilson MM, Wang C, Selby M, Taube JM, Anders R, Chen L, Korman AJ, Pardoll

DM, Lowy I, Topalian SL (2010) Phase I study of single-agent anti-programmed death-1 (MDX-1106) in

refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. Journal

of Clinical Oncology, 2010 Jul 1; 28(19): 3167-3175. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J,

Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF,

Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM (2012) Safety

and activity of anti-PD-L1 antibody in patients with advanced cancer. New England Journal of Medicine,

2012 Jun 28; 366(26): 2455-2465. Brunet JF, Denizot F, Luciani MF, Roux-Dosseto M, Suzan M, Mattei MG, Golstein P (1987) A new

member of the immunoglobulin superfamily: CTLA-4. Nature, 1987 Jul 16-22; 328(6127): 267-270. Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, Fong L (2014) Improved survival with T

cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Science Translational Medicine,

2014 May 28; 6(238): 238ra70.

Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, Kuo F, Kendall SM, Requena

D, Riaz N, Greenbaum B, Carroll J, Garon E, Hyman DH, Zehir A, Solit D, Berger M, Zhou R, Rizvi

NA, Chan TA (2018) Patient HLA class 1 genotype influences cancer response to checkpoint blockade

immunotherapy. Science, 2018 Feb 2; 359: 582-587.

De Martin E, Michot JM, Papouin B, Champiat S, Mateus C, Lambotte O, Roche B, Antonini TM, Coilly

A, Laghouati S, Robert C, Marabelle A, Guettier C, Samuel D (2018) Characterization of liver injury

induced by cancer immunotherapy using immune checkpoint inhibitors. Journal of Hepatology, 2018 Jun;

68(6): 1181-1190. doi: 10.1016/j.jhep.2018.01.033.

Eggermont AM, Chiarion-Sileni V, Grob JJ, Dummer R, Wolchok JD, Schmidt H, Hamid O, Robert C,

Ascierto PA, Richards JM, Lebbé C, Ferraresi V (2016) Prolonged Survival in Stage III Melanoma with

Ipilimumab Adjuvant Therapy. New England Journal of Medicine, 2016 Nov 10; 19(375):1845-1855.

Eroglu Z, Kim DW, Wang X, Camacho LH, Chmielowski B, Seja E, Villanueva A, Ruchalski K, Glaspy

JA, Kim KB, Hwu WJ, Ribas A (2015) Long term survival with cytotoxic T lymphocyte-associated

antigen 4 blockade using tremelimumab. European Journal of Cancer, 2015 Nov; 51(17): 2689-2697.

Frankel JC (2017) Autoimmune diseases surface after cancer treatment. Biomedicine, 2017 Nov 17;

358(6365): 852.

Page 92: CANCER VACCINES - Worcester Polytechnic Institute

92

Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki

T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR,

Honjo T (2000) Engagement of the PD-1 immuno-inhibitory receptor by a novel B7 family member leads

to negative regulation of lymphocyte activation. Journal of Experimental Medicine, 2000 Oct 2; 192(7):

1027-1034. Garber K (2018) A New Cancer Immunotherapy Suffers a Setback. Science, 2018 May 11; 360(6389):

588.

Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, Powderly JD, Heist RS, Carvajal

RD, Jackman DM, Sequist LV, Smith DC, Leming P, Carbone DP, Pinder-Schenck MC, Topalian SL,

Hodi FS, Sosman JA, Sznol M, McDermott DF, Pardoll DM, Sankar V, Ahlers CM, Salvati M,

Wigginton JM, Hellmann MD, Kollia GD, Gupta AK, and Brahmer JR (2015) Overall Survival and

Long-Term Safety of Nivolumab (Anti-Programmed Death-1 Antibody, BMS-936558, ONO-4538) in

Patients with Previously Treated Advanced Non-Small-Cell Lung Cancer. Journal of Clinical Oncology,

0.1200/JCO.2014.58.3708.

Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets CV, Prieto PA, Vicente D,

Hoffman K, Wei SC, Cogdill AP, Zhao L, Hudgens CW, Hutchinson DS, Manzo T, Macedo MP,

Cotechini T, Kumar T, Chen W, Reddy SM, Sloane RS, Pena JG, Jiang H (2018) Gut microbiome

modulates response to anti-PD-1 immunotherapy in melanoma patients. Science, 2018 Jan 5: 359(6371):

97-103. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber

JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A,

Mateus C, Boasberg P, Tumeh PC, Chmielowski B, Ebbinghaus SW, Li XN, Kang SP, Ribas A (2013)

Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. New England Journal of

Medicine, 2013 Jul 11; 369(2): 134-144. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF,

Powderly JD, Gettinger SN, Kohrt HE, Horn L, Lawrence DP, Rost S, Leabman M, Xiao Y, Mokatrin A,

Koeppen H, Hegde PS, Mellman I, Chen DS, Hodi FS (2014) Predictive correlates of response to the anti-

PD-L1 antibody MPDL3280A in cancer patients. Nature, 2014 Nov 27; 515(7528): 563-567. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G,

Tamada K, Chen L (2005) Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer

therapeutic immunity. Cancer Research, 2005 Feb 1; 65(3): 1089-1096. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, Davis T, Henry-Spires R, MacRae

S, Willman A, Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman A, Allison JP, Dranoff G (2003)

Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously

vaccinated metastatic melanoma and ovarian carcinoma patients. Proceedings of the National Academy

of Sciences USA, 2003 Apr 15; 100(8): 4712-4717.

Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C,

Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette

GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J,

Yellin MJ, Nichol GM, Hoos A, Urba WJ (2010) Improved survival with ipilimumab in patients with

metastatic melanoma. New England Journal of Medicine, 2010 Aug 19; 363(8): 711-723.

Jobin C (2018) Precision medicine using microbiota. Science, 2018 Jan 5; 359(6371): 32-34.

Page 93: CANCER VACCINES - Worcester Polytechnic Institute

93

Kaiser J (2018) Too much of a good thing? Science, 2018 Mar 23; 359(6382): 1346-1347.

doi: 10.1126/science.359.6382.1346.

Ledford H (2018) Promising cancer drug hits snags. Nature, 2018 Apr 12; 556: 161-162.

Lee B, Mukhi N, Liu D (2012) Current management and novel agents for malignant melanoma. Journal

of Hematology and Oncology, 2012 Feb 14; 5(3): 2-7.

Lussier DM, O'Neill L, Nieves LM, McAfee MS, Holechek SA, Collins AW, Dickman P, Jacobsen J,

Hingorani P, Blattman JN (2015) Enhanced T-cell immunity to osteosarcoma through antibody blockade

of PD-1/PD-L1 interactions. Journal of Immunotherapy, 2015 Apr; 38(3): 96-106. Lussier DM, Johnson JL, Hingorani P, Blattman JN (2015) Combination immunotherapy with α-CTLA-4

and α-PD-L1 antibody blockade prevents immune escape and leads to complete control of metastatic

osteosarcoma. Journal for Immunotherapy of Cancer, 2015 May 19; 3: 21.

McCune JS (2018) Rapid Advances in Immunotherapy to Treat Cancer. Clinical Pharmacology and

Therapeutics, 2018 April; 103(4): 540-544. Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L (2007) Immuno-stimulatory monoclonal

antibodies for cancer therapy. Nature Reviews Cancer, 2007 Feb; 7(2): 95-106.

Motzer RJ, Tannir NM, McDermott DF, Frontera OA, Melichar B, Choueiri TK, Plimack ER,

Barthélémy P, Porta C, George S, Powles T, Donskov F (2018) Nivolumab plus Ipilimumab versus

Sunitinib in Advanced Renal-Cell Carcinoma. New England Journal of Medicine, 2018 Apr 5; 378(14):

1277-1290. National Cancer Institute (2016) FDA Approves Atezolizumab for Bladder Cancer.

https://www.cancer.gov/news-events/cancer-currents-blog/2016/fda-atezolizumab-bladder

Peggs KS, Quezada SA, Korman AJ, Allison JP (2006) Principles and use of anti-CTLA4 antibody in

human cancer immunotherapy. Current Opinion in Immunology, 2006 Apr; 18(2): 206-213.

Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, Linette GP, Meyer N,

Giguere JK, Agarwala SS, Shaheen M, Ernstoff MS (2015) Nivolumab and Ipilimumab versus

Ipilimumab in Untreated Melanoma. New England Journal of Medicine, 2015 May 21; 21(372): 2006-

2017.

Ravi R, Noonan KA, Pham V, Bedi R, Zhavoronkov A, Ozerov IV, Makarev E, Artemov AV, Wysocki

PT, Mehra R, Nimmagadda S, Marchionni L, Sidransky D, Borrello IM, Izumchenko E, Bedi A (2018)

Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFβ

enhance the efficacy of cancer immunotherapy. Nature Communications, 2018; 9(1): 741.

Ribas A, Camacho LH, Lopez-Berestein G, Pavlov D, Bulanhagui CA, Millham R, Comin-Anduix B,

Reuben JM, Seja E, Parker CA, Sharma A, Glaspy JA, Gomez-Navarro J (2005) Antitumor activity in

melanoma and anti-self responses in a phase I trial with the anti-cytotoxic T lymphocyte-associated

antigen 4 monoclonal antibody CP-675,206. Journal of Clinical Oncology, 2005 Dec 10; 23(35): 8968-

8977.

Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, Joshua AM, Patnaik A, Hwu WJ, Weber

JS, Gangadhar TC, Hersey P, Dronca R, Joseph RW, Zarour H, Chmielowski B, Lawrence DP, Algazi A,

Rizvi NA, Hoffner B, Mateus C, Gergich K, Lindia JA, Giannotti M, Li XN, Ebbinghaus S, Kang SP,

Page 94: CANCER VACCINES - Worcester Polytechnic Institute

94

Robert C (2016) Association of Pembrolizumab With Tumor Response and Survival Among Patients

With Advanced Melanoma. JAMA, 2016 Apr 19; 315(15): 1600-1609. doi: 10.1001/jama.2016.4059.

Ribas A, Wolchok JD (2018) Cancer immunotherapy using checkpoint blockade. Science, 2018 Mar 23:

359: 1350-1355. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS,

Miller ML, Rekhtman N, Moreira AL, Ibrahim F, Bruggeman C, Gasmi B, Zappasodi R, Maeda Y,

Sander C, Garon EB, Merghoub T, Wolchok JD, Schumacher TN, Chan TA (2015) Cancer immunology.

Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science,

2015 Apr 3; 348(6230): 124-128. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, Weber JS, Joshua AM, Hwu WJ,

Gangadhar TC, Patnaik A, Dronca R, Zarour H, Joseph RW, Boasberg P, Chmielowski B, Mateus C,

Postow MA, Gergich K, Elassaiss-Schaap J, Li XN, Iannone R, Ebbinghaus SW, Kang SP, Daud A

(2014) Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory

advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet, 2014 Sep 20;

384(9948): 1109-1117. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C,

Kalinka-Warzocha E, Savage KJ, Hernberg MM, Lebbé C, Charles J, Mihalcioiu C, Chiarion-Sileni V,

Mauch C, Cognetti F, Arance A, Schmidt H, Schadendorf D, Gogas H, Lundgren-Eriksson L, Horak C,

Sharkey B, Waxman IM, Atkinson V, Ascierto PA (2015) Nivolumab in previously untreated melanoma

without BRAF mutation. New England Journal of Medicine, 2015 Jan 22; 372(4): 320-330.

Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N,

O'Donnell PH, Balmanoukian A, Loriot Y, Srinivas S, Retz MM, Grivas P, Joseph RW, Galsky MD,

Fleming MT, Petrylak DP, Perez-Gracia JL, Burris HA, Castellano D, Canil C, Bellmunt J, Bajorin D,

Nickles D, Bourgon R, Frampton GM, Cui N, Mariathasan S, Abidoye O, Fine GD, Dreicer R (2016)

Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed

following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet,

2016 May 7; 387(10031): 1909-1920. doi: 10.1016/S0140-6736(16)00561-4.

Routy B, Chatelier EL, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M,

Rauber C, Roberti MP, Fidelle M, Flament C, Colame VP, Opolon P, Klein C, Iribarren K, Mondragón L,

Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, Masip JR, Naltet C (2018) Gut microbiome

influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science, 2018 Jan 5;

359(6371): 91-97.

Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, Patt D, Chen TT, Berman DM,

Wolchok JD (2015) Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of

Ipilimumab in Unresectable or Metastatic Melanoma. Journal of Clinical Oncology, 2015 Jun 10; 33(17):

1889-1894.

Sharma P, Allison JP (2015) The future of immune checkpoint therapy. Cancer Immunology and

Immunotherapy, 2015 Apr 3; 348(6230): 56-61. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong

P, Ho TS, Hollmann TJ, Bruggeman C, Kannan K, Li Y, Elipenahli C, Liu C, Harbison CT, Wang L,

Ribas A, Wolchok JD, Chan TA (2014) Genetic basis for clinical response to CTLA-4 blockade in

melanoma. New England Journal of Medicine, 2014 Dec 4; 371(23): 2189-2199.

Page 95: CANCER VACCINES - Worcester Polytechnic Institute

95

Sznol M, Chen L (2013) Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced

human cancer. Clinical Cancer Research, 2013 Mar 1; 19(5): 1021-1034. Tanner L (2015) More Cancer Success with Drugs that Enlist Immune System. Worcester Telegram and

Gazette, Issue 30 May 2015.

Tauriello DVF, Ponce SP, Stork D, Llergo AB, Ramentol JB, Iglesias M, Sevillano M, Ibiza S, Cañellas

A, Momblona XH, Byrom D, Matarin JA, Calon A, Rivas EI, Nebreda AR, Riera A, Attolini CSO, Batlle

E (2018) TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature, 2018

Feb 22; 554: 538-568.

Topalian SL, Drake CG, Pardoll DM (2012) Targeting the PD-1/B7-H1 (PD-L1) pathway to activate anti-

tumor immunity. Current Opinion in Immunology, 2012 Apr; 24(2): 207-212. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal

RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen

L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S,

McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M (2012) Safety, activity, and immune

correlates of anti-PD-1 antibody in cancer. New England Journal of Medicine, 2012 Jun 28; 366(26):

2443-2454. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, Brahmer JR, Lawrence

DP, Atkins MB, Powderly JD, Leming PD, Lipson EJ, Puzanov I, Smith DC, Taube JM, Wigginton JM,

Kollia GD, Gupta A, Pardoll DM, Sosman JA, Hodi FS (2014) Survival, durable tumor remission, and

long-term safety in patients with advanced melanoma receiving nivolumab. Journal of Clinical Oncology,

2014 Apr 1; 32(10): 1020-1030. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, Thompson CB, Bluestone

JA (1994) CTLA-4 can function as a negative regulator of T cell activation. Immunity, 1994 Aug; 1(5):

405-413. Weber JS, Kähler KC, Hauschild A (2012) Management of immune-related adverse events and kinetics of

response with ipilimumab. Journal of Clinical Oncology, 2012 Jul 20; 30(21): 2691-2697. Weber JS, Kudchadkar RR, Yu B, Gallenstein D, Horak CE, Inzunza HD, Zhao X, Martinez AJ, Wang

W, Gibney G, Kroeger J, Eysmans C, Sarnaik AA, Chen YA (2013) Safety, efficacy, and biomarkers of

nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. Journal of Clinical Oncology,

2013 Dec 1; 31(34): 4311-4318. Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH

Jr, Lao CD, Linette GP, Thomas L, Lorigan P, Grossmann KF, Hassel JC, Maio M, Sznol M, Ascierto

PA, Mohr P, Chmielowski B, Bryce A, Svane IM, Grob JJ, Krackhardt AM, Horak C, Lambert A, Yang

AS, Larkin J (2015) Nivolumab versus chemotherapy in patients with advanced melanoma who

progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3

trial. Lancet Oncology, 2015 Apr; 16(4): 375-384. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE,

Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I,

Inzunza HD, Feely W, Horak CE, Hong Q, Korman AJ, Wigginton JM, Gupta A, Sznol M (2013)

Nivolumab plus ipilimumab in advanced melanoma. New England Journal of Medicine, 2013 Jul 11;

369(2): 122-133.

Page 96: CANCER VACCINES - Worcester Polytechnic Institute

96

Wolchok JD, Sileni VC, Gonzalez R, Rutkowski P, Grob JJ, Cowey CJ, Lao CD, Wagstaff J,

Schadendorf D, Ferrucci PF, Smylie MS, Dummer R (2017). Overall Survival with Combined Nivolumab

and Ipilimumab in Advanced Melanoma. New England Journal of Medicine, 2017 Oct 5; 377(14): 1345-

1356.

Zarrabi K, Wu S (2018) Risk of Liver Toxicity with Nivolumab Immunotherapy in Cancer Patients.

Oncology, 2018; 94(5): 259-273. doi: 10.1159/000486679.

Page 97: CANCER VACCINES - Worcester Polytechnic Institute

97

METHODS

To accomplish project objective-1, we performed an extensive review of the available research

literature, including reputable academic journal articles, relevant books, scholarly websites, newspaper

articles, and other pertinent materials.

To accomplish objective-2, we conducted a set of interviews with various biomedical

researchers who have performed cancer vaccine research on either animals or humans. We also

interviewed scientists who have used traditional cancer fighting technologies, to determine their range of

opinions on newer forms of treatment.

Who: The “stakeholders” (interviewees) included academic biomedical experts on cancer

vaccines and traditional cancer therapies, and general health experts on cancer. These experts helped

answer questions resulting from our Lit Review search, and helped us prioritize any remaining problems.

Some of the stakeholders interviewed were initially identified by referral from the project advisor, Prof.

David Adams. Other interviewees were identified from the published literature as authors on key

scientific papers, or were referred to us from the initial interviewees.

Where and When: Once contact was made with a potential interviewee (see below), a time and

place was set up for the interview to be performed at the interviewee’s workplace. Whenever possible,

interviews were conducted in person, although most were conducted by email, phone, or Skype.

How: Our first round of prospective interviewees was contacted by email and/or phone. If no

response was received, we used follow-up emails and phone calls. We developed our interview questions

(see preliminary questions in the Appendix) based on our review of the literature, and tailored the

questions to the interviewee’s expertise. Based on the interviewee’s response to our first questions, in

some cases we asked follow-up questions to clarify the information provided, or to press the interviewee

for more information. The preliminary list of questions in the Appendix covers the full range of topics

needed to cover our project.

With respect to the method of the interview, whenever possible each interview involved two team

members, so that one member could ask questions while the other member wrote detailed notes, and vice

versa. We asked whether the interviewee consented to be digitally recorded, and if not, we used written

notes or emails as the main method of recording the conversation.

At the start of the interview, we informed the interviewee about the purpose of our project, and

asked for permission to quote them (see draft interview preamble in the Appendix). We explained how we

will protect their confidentiality, if necessary, by giving them the right to review any quotations used in

the final published report, explaining that the interview is voluntary, and explaining that they may stop the

interview at any time or refuse to answer any question.

After the interview, we asked each interviewee for permission to follow-up with them at a later

date if needed to fill in any gaps in the information. And, as mentioned above, we asked the interviewee

to recommend other potential stakeholders we might interview, to further increase the number of

interviews with key individuals.

With respect to the total number of interviews needed for our project, we stopped interviewing

additional subjects when we obtained a sufficient amount of information to represent all sides of the

cancer vaccine story, good and bad, and when all unclear points had been clarified.

Page 98: CANCER VACCINES - Worcester Polytechnic Institute

98

To accomplish objectives-3 and 4, the group synthesized all of the information collected in the

literature research, interviews, and follow-up interviews, to ascertain the strength of the evidence, and to

create recommendations for further research.

Page 99: CANCER VACCINES - Worcester Polytechnic Institute

99

RESULTS / FINDINGS

Our review of the cancer vaccine literature identified several problems that needed further

clarification in interviews. We performed interviews with a variety of scientists, including those who

helped develop cancer vaccines, doctors who performed cancer vaccine clinical trials, and scientists

actively engaged in developing new generations of cancer vaccine drugs. We chose to focus on two main

problems and directions: 1) What causes vaccine side-effects, and what new approaches might minimize

them? 2) Why are some patients resistant to vaccines, and what new approaches might improve this?

Results for Therapeutic Antibody Vaccines A major problem with cancer vaccines is that a substantial proportion of the patient’s tumors do

not respond to them. To understand this problem further, we interviewed Dr. Sergey E. Sedykh of the

Laboratory of Repair Enzymes, Siberian Branch of the Russian Academy of Sciences Institute of

Chemical Biology and Fundamental Medicine, Novosibirsk State University, Novosibirsk, Russia. Dr.

Sedykh was corresponding author on a 2018 review article on bi-specific antibodies: Sedykh et al., 2018,

Bispecific antibodies: design, therapy, perspectives. Drug Design, Development, and Therapy, 2018 Jan

22; 12: 195-208. When asked his opinion on the leading cause of resistance to bi-specific antibody

therapies, he replied: “My opinion is that bispecific antibodies (like blinatumomab and catumaxomab) act

by attracting a T-cell (via the CD3 receptor) to a tumor cell, but if there are few T-cells remaining in the

patient [for example due to a prior chemotherapy], the patient cannot fight the cancer. The down-

regulation of surface antigens on cancer cells may be another tumor-evolution route of cancer cell

resistance. In some cases, therapeutic BsAbs are considered as the auxiliary treatment after inductional

and consolidation therapy”. So, Dr. Sedykh indicated that tumor resistance to bi-specific antibody

therapy can result from a general low number of T-cells to attract to the tumor (with the anti-CD3

portion), and from the down-regulation of target antigen expression.

Our Lit Review indicated that one approach for overcoming resistance is to change the

type of vaccine used. A very good example of this is the paper: Rothe et al., 2015, A phase-I study

of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with relapsed or refractory

Hodgkin lymphoma. Blood, 2015 Jun 25; 125(26): 4024-4031. The authors used bispecific antibody

AFM13, which has affinities for CD30 (present in lymphomas) and CD16A (which recruits natural killer

cells to the tumor instead of T-cells). This was a Phase-I dose-escalation study (doses of 0.01 to 7 mg/kg

body weight) of 28 patients with heavily pretreated relapsed or refractory Hodgkin lymphoma. Three of

26 evaluable patients (11.5%) achieved partial remission, and 13 patients (50%) achieved stable disease.

Importantly, AFM13 was active in brentuximab vedotin (CD30)-refractory patients. A phase 2 study is

currently planned. Adverse events were generally mild to moderate. So, attracting NK cells to a tumor

might be an alternative approach for treating patients resistant to T-cell attracting therapies. To shed

more light on this topic, we interviewed the corresponding author of the article: Dr. Andreas

Engert, Professor of Internal Medicine, Hematology & Oncology, and Chairman of the German Hodgkin

Study Group, Department of Internal Medicine, University Hospital of Cologne, Germany. When asked

whether their NK-recruiting strategy to switch the method of cell killing might work for other types of

cancers besides Kodgkin lymphoma, he stated: “We have only used this approach in Hodgkin lymphoma,

but it might certainly be possible that this also works in other malignancies, we have not pursued this

yet”. Thus, Dr. Engert indicated that this new approach of switching the method of cell killing might

indeed work with other types of cancers, but they have not yet tested it. Another possible mechanism for tumor resistance is a mutation occurs that switches the tumor

from one growth signaling pathway to another. An example of this is the article: Lieu et al., 2017, A

Page 100: CANCER VACCINES - Worcester Polytechnic Institute

100

Phase Ib Dose-Escalation Study of the Safety, Tolerability, and Pharmacokinetics of Cobimetinib and

Duligotuzumab in Patients with Previously Treated Locally Advanced or Metastatic Cancers with Mutant

KRAS. Oncologist, 2017 Sep; 22(9): 1024-e89. The authors investigated patients with KRAS-mutant

tumors that possess abnormal MAPK pathway signaling and cell proliferation. They did a Phase-IB dose-

escalation study of a combination of Cobimetinib (which blocks MAPK signaling) and the bi-specific

antibody duligotuzumab (which inhibits ligand binding to two types of receptors: EGFR and human

epidermal growth factor receptor 3 (HER3). They enrolled 23 patients KRAS-mutant tumors. Nine of

the 23 patients (39%) showed stable disease, but 14 were non-responders. To get at the basis of the tumor

resistance, we interviewed the corresponding author on the article: Dr. Christopher H. Lieu, MD,

Director of GI Medical Oncology, and Deputy Associate Director for Clinical Research, University of

Colorado Anschutz Medical Campus, Division of Medical Oncology, Aurora, Colorado. When asked his

opinion about why the patients were non-reponders, he replied: “I think if a patient’s tumor is not

addicted to the MAPK pathway, they may be using another growth pathway to grow and metastasize. In

our case, the non-responders may not have needed the MAPK pathway (which both of our drugs inhibit),

but may have used another growth pathway that we were not blocking”. Thus, according to Dr. Lieu,

when using therapies that work by binding to receptors to block a specific cell growth pathway, to

determine whether a particular tumor will respond to the antibody, the tumor needs to be verified as being

dependent on that specific pathway, otherwise the treatment won’t work.

Reduction of target antigen expression in a portion of the tumor is another mechanism of

resistance. This situation was seen in a recent 2018 paper: Bosco et al., 2018, Preclinical evaluation of a

GFRA1-targeted antibody-drug conjugate in breast cancer. Oncotarget, 2018 May 1; 9(33): 22960-22975.

The authors used a genomics approach to identify membrane-localized tumor-associated antigens (TAAs)

in breast cancer cells that might serve as a target for antibody-drug conjugate (ADC) therapy. They

identified glial cell line-derived neurotrophic factor (GDNF) family receptor-α1 (GFRA1) as a breast

cancer tumor-associated antigen. They determined that GFRA1 shows limited expression in normal cells,

over-expression in some breast tumor subtypes, and rapid internalization, making it a good ADC target.

Their GFRA-targeting ADC showed strong anti-tumor activity against GFRA1-positive tumor cell lines

in vitro, and in vivo against patient-derived human cancers in mouse xenograft models. The safety profile

showed only transient problems in the bone marrow and peripheral blood, consistent with well known

off-target effects of their chosen cytotoxic cargo. To determine the proportion of breast cancer patients

that might benefit from anti-GFRA therapy, we interviewed the corresponding author of the paper: Dr.

Emily E. Bosco, PhD, Scientist-II, Oncology Research, MedImmune, Gaithersburg, Maryland. When

asked her opinion of what percent of breast cancer patients might express GFRA and benefit from their

ADC, she replied: “we would first treat the moderate and strong GFRA-expressing breast cancer patients,

and based on immunohistochemistry (IHC) these patients represent about 20% of estrogen receptor-

positive breast cancer patients, and about 9% of triple negative breast cancer patients”. So, Dr. Bosco

indicates that based on their own immunohistochemical staining of breast cancer tumors, about 29% of

breast cancer patients might benefit from the GFRA-targeting ADC drug. Presumably, the other breast

cancer patients would need to be treated with a drug targeting a different surface antigen, such as HER2

or HER3.

A problem frequently encountered with new therapies is a lack of negative controls in the clinical

trials. In the case of cancer vaccines, the patients are not allowed to receive the new therapy unless the

cancer relapses from the patient’s previous treatments. And the prognosis is very poor for relapsed

patients, so almost any increase in patient survival is significant for these populations. An example of this

problem is the article: Trněný et al., 2018, A Phase 2 multicenter study of the anti-CD19 antibody drug

conjugate coltuximab ravtansine (SAR3419) in patients with relapsed or refractory diffuse large B-cell

lymphoma previously treated with rituximab-based immunotherapy. Haematologica, 2018 May 10. The

authors performed a Phase-II multi-center clinical trial on the safety and efficacy of their anti-CD19-

targeting antibody-drug conjugate (ADC) Coltuximab ravtansine. They analyzed patients with relapsed

or refractory diffuse large B-cell lymphoma who had previously received Rituximab therapy (antibody

Page 101: CANCER VACCINES - Worcester Polytechnic Institute

101

targeting CD20). 41 patients were included in the treatment population. The overall response rate was

18/41 (43.9%). The median duration of response, progression-free survival, and overall survival were: 4.7

months, 4.4 months, and 9.2 months, respectively. Common non-hematologic adverse events included

asthenia/fatigue (30%), nausea (23%), and diarrhea (20%). Grade 3-4 adverse events were reported in 23

patients (38%), the most frequent being hepatotoxicity (3%) and abdominal pain (3%). In this trial, there

was no patient cohort treated only with traditional chemotherapy (negative control) because all patients

were resistant to traditional treatments and received the ADC treatment. We interviewed the

corresponding author of the paper Dr. Marek Trněný, MD, CSc, Professor and Chairman, 1st Dept

Medicine, Charles University, General Hospital, Praha, Czech Republic. When asked his opinion about

the median overall survival rate for the non-ADC-treated patients, he responded: “Generally speaking,

their median overall survival is about one year, but some patients could reach long-term survival”. So,

Dr. Trněný indicates that for the patients they used for the Phase-II tests (with relapsed or refractory

diffuse large B-cell lymphoma who had previously received Rituximab therapy), the median overall

survival is about 1 year. This is slightly more than the 9.2 months seen for their patients treated with their

ADC drug, so the ADC did not appear to lengthen overall survival in this particular study. This example

shows the difficulty of determining whether a drug benefits a population whose prognosis is extremely

poor from the beginning of the study.

Results for Dendritic Cell Vaccines In the cancer vaccine field, the experiments typically move from pre-clinical testing (which

includes testing the drug against cancer cell lines in vitro, and testing xenograft mice in vivo) into human

clinical trials. But it is not clear exactly how much information is needed from the pre-clinical testing

before moving into clinical trials. For example, we identified the following paper: Choi et al., 2018,

Combination Treatment of Stereotactic Body Radiation Therapy and Immature Dendritic Cell

Vaccination for Augmentation of Local and Systemic Effects. Cancer Research and Treatment, 2018 Jun

6. doi: 10.4143/crt.2018.186. The authors investigated dendritic cell (DC) vaccines and methods for

improving the efficiency of priming the DCs with tumor antigens. DCs can be primed against tumor

antigens in vivo in the patient, or ex-vivo by mixing isolated DCs with tumor antigens outside the

body. The ex-vivo approach is used most often in clinical trials, but the in vivo method has the advantage

of being available even when no tumor biopsy tissue is available to do the ex-vivo priming. Ionizing

radiation has recently been tested as a method for facilitating in vivo DC antigen priming, but exactly how

the radiation works to prime the DC cells is not clear. The radiation might help release tumor associated

antigens from the tumor, or it might increase the release of damage-associated molecular patterns

(DAMPs) (such as heat shock proteins) from dying tumor cells. In the Choi et al., 2018 study, the authors

investigated the use of stereotactic body radiation therapy (SBRT) as a method for facilitating the

presentation of tumor-associated antigens (TAA) to immature dendritic cells (iDCs). They tested their

method on mouse xenograft models of CT-26 colon carcinoma with 3 groups of mice: 1) radiation

therapy alone, 2) intra-tumor injection of DCs electroporated with tumor antigens, or 3) the combination

treatment. The data showed that the radiation method achieved the best DC priming and T-cell activation

compared to other priming methods, and that mouse survival was highest when using the combination

treatment. The authors conclude that clinical trials are warranted. We interviewed the corresponding

author on the paper: Dr. Chul Won Choi, MD, Department of Radiation Oncology, Dongnam Institute of

Radiological & Medical Sciences, Busan, Korea. When asked his opinion about whether it will be

necessary to more deeply understand the mechanism of how the radiation is facilitating the DC antigen

presentation process before proceeding to clinical trials, he responded: “Actually, I don't think so. There

are already many suggested mechanisms for how radiation enhances immune reactions. However, doing a

study on the ratio of apoptosis [programmed cell death] to necrosis [generalized cell death] after

irradiation would be interesting. Knowing this ratio would be helpful for determining the dose of

radiation, or for enhancing efficacy”. When asked whether his team was moving forward with clinical

Page 102: CANCER VACCINES - Worcester Polytechnic Institute

102

trials, he responded: “No. There are many limitations to moving to clinical trials including financial,

administrative, and legal issues. So, we won’t be going to the next step”. So, Dr. Choi thinks that it likely

is not necessary to obtain a full understanding of exactly how their radiation method works to prime DC

cells before proceeding to clinical trials, but that his lab is not moving forward due to financial,

administrative, legal, and scientific reasons.

As mentioned previously, for some of the clinical trials published in the literature, it was not clear

how the patient prognoses for patients receiving the cancer vaccine fared relative to untreated patients, or

to patients treated with traditional chemotherapy, because all patients receiving cancer vaccines have

received previous therapies and have relapsed/recurring cancers. For example, we identified the following

paper: Liau et al., 2018, First results on survival from a large Phase 3 clinical trial of an autologous

dendritic cell vaccine in newly diagnosed glioblastoma. Journal of Translational Medicine, 2018 May 29;

16(1): 142. This paper showed the results of a Phase-III trial of 331 patients with glioblastoma. The

standard therapy for glioblastomas is surgery, radiotherapy, and oral chemotherapy temozolomide. This

study evaluated the addition of an autologous tumor lysate-pulsed dendritic cell vaccine (DCVax®-L) to

standard therapy for newly diagnosed glioblastoma patients. After surgery and chemo-radiotherapy,

patients were randomly assigned to two groups: 1) chemo + DC vaccine (232 patients), or 2) chemo +

placebo (99 patients). However, if the patient’s cancer recurred, the patient was allowed to receive the DC

vaccine regardless of the initial group assignment. So, because of this “cross-over design”, nearly 90% of

the patients received the DC vaccine, and patient survival relative to a placebo could not be determined.

The median overall survival was 34.7 months, with a 3-year survival of 46.4%. We interviewed the

corresponding author on the paper: Dr. Linda M. Liau, MD, of the University of California Los Angeles

(UCLA) David Geffen School of Medicine & Jonsson Comprehensive Cancer Center, Los Angeles, CA.

When asked whether the median survival of 34.7 months was significantly better than the placebo group,

she responded: “Yes, the median overall survival for the entire intent-to-treat (ITT) population does seem

longer than the normal prognosis for glioblastoma patients based on historical controls” So, Dr. Liau

believes the average survival for the patients receiving her dendritic cell vaccine is longer than patients

not receiving it. Because most of the patients in her cross-over study received the DC vaccine, we must

rely on "historical data" for the untreated patients.

One trend observed in the literature is the use of combination cancer vaccines. Research teams

that had previously investigated single cancer vaccines were now testing combinations. And teams that

had previously tested combinations were now expanding the combinations to other types of cancer. As an

example, DC vaccines are being combined with checkpoint inhibitor vaccines. Checkpoint vaccines are

used to over-ride the immuno-suppression caused by the tumor cell interacting with the immune system

(typically DCs and T-cells). Three checkpoint targets came up repeatedly in our review: PD-1, PD-L1,

and CTLA-4. But given their importance, it makes sense to identify new checkpoint receptors as vaccine

targets. An interesting system was recently established using tumor “exosomes” (membrane vesicles) to

identify checkpoint inhibitors: Ning et al., 2018, Tumor exosomes block dendritic cells maturation to

decrease the T cell immune response. Immunology Letters, 2018 Jul; 199: 36-43. In this study, the authors

developed a system for determining whether exosomes produced by two particular types of cancer (LLC

Lewis lung carcinoma or 4T1 breast cancer) contribute to DC suppression. They found that exosomes

from these tumors indeed blocked the differentiation of myeloid precursor cells into DC cells, and

inhibited the migration and maturation of DCs. In addition, the inhibitory response was partially blocked

by treating with anti-PD-L1 antibody, suggesting this PD-L1 checkpoint inhibition is important to the

inhibition. We interviewed the corresponding author of the paper: Dr. Chunjian Qi, MD, PhD,

Professor and Director, Medical Research Center, The Affiliated Changzhou No.2 People's Hospital of

Nanjing Medical University, Changzhou, China. When asked now that he had set up the exosome system

for studying the inhibition process, whether he intends to investigate other potential blockers of the DC

activation besides PD-L1, he responded: “While PD-L1 antibody worked well, other blockers were not

investigated in this exosome system”. So, Dr. Qi indicated that they have not yet investigated any other

Page 103: CANCER VACCINES - Worcester Polytechnic Institute

103

checkpoint blockers (such as CTLA-4), so the latter could in theory also be involved in the inhibition

process.

An example of a new therapy that could in theory be combined with another to make a

combination is the following paper: Benitez-Ribas et al., 2018, Immune Response Generated With the

Administration of Autologous Dendritic Cells Pulsed With an Allogenic Tumoral Cell-Lines Lysate in

Patients With Newly Diagnosed Diffuse Intrinsic Pontine Glioma. Frontiers in Oncology, 2018 Apr 26;

8: 127. This was a Phase-Ib clinical trial of 9 patients with diffuse intrinsic pontine glioma (DIPG), a

lethal brainstem tumor in children. The authors tested autologous dendritic cell vaccines (ADCVs) pulsed

with allogeneic tumor cell-line lysates in newly diagnosed patients following radiation therapy. The DCs

were prepared from monocytes obtained by leukapheresis. The authors found that their pulsing procedure

boosted non-specific immune responses (KLH) in 9 of 9 patients, and it boosted specific anti-tumor

immune responses in 8 of 9 patients, in both PBMCs and T-lymphocytes isolated from CSF. We

interviewed one of the two corresponding authors on the paper: Dr. Andrés Morales La Madrid, MD,

Unidad de Neuro Oncología Pediátrica, Servicio de Oncología y Hematología Pediátrica, Hospital St Joan

de Déu, Passeig St Joan de Déu, Barcelona, Spain. When asked his opinion whether his pulsed dendritic

cell vaccine approach might be promising for use with other types of immunotherapies (especially

combined with checkpoint vaccines), he responded: “Based on the unpublished data from our lab we are

considering combining our DC vaccines with CTLA-4 inhibitors”. So, indeed he agreed with our

assessment that his DC vaccine approach for childhood glioma tumors might work better when combined

with a checkpoint vaccine (in his case combined with anti-CTLA-4).

For DC vaccines, using the ex vivo priming method usually involves mixing immature DC cells

with a tumor lysate. But when priming the DC cells in vivo in a patient, it is not clear how the DC cells

are primed. One example of this is the article: Garzon-Muvdi et al., 2018, Dendritic cell activation

enhances anti-PD-1 mediated immunotherapy against glioblastoma. Oncotarget, 2018 Apr 17; 9(29):

20681-20697. Glioblastoma (GBM) tumors strongly suppress the immune system. Checkpoint blockage

vaccines (such as antibodies against PD-1) might be useful for overcoming the blockade, but some

experiments suggest the checkpoint approach may not be sufficient by itself. The authors investigated the

activation of DC cells as a supplement to anti-PD-1 therapy in mice. Their data shows that activating

DCs by stimulating the TLR3 receptor with poly(I:C) enhances the PD-1 anti-tumor response, and

increases survival in mouse glioblastoma models. DC-depletion experiments showed that DCs are

required for the anti-tumor response. We interviewed the corresponding author for the paper: Dr.

Michael Lim, MD, Professor of Neurosurgery, Oncology, Radiation Oncology, Otolaryngology, and

Institute of NanoBiotechnology, Director of the Brain Tumor Immunotherapy Program, Director of the

Metastatic Brain Tumor Center, Johns Hopkins University School of Medicine, Baltimore, MD. When

asked about moving forward with clinical trials and whether he plans on using poly(I:C) to activate the

patient’s DC cells, he responded: “Yes, we are considering using polyIC”. So, Dr. Lim believes that

activating the toll-like receptor-3 TLR-3 using poly(I:C) should work in human patients, and can

supplement his anti-PD-1 therapy.

Results for TIL Vaccines

As mentioned previously, our review of the literature indicated that using combinations of

vaccines is a strong trend in the cancer vaccine field. An example of this using tumor-infiltrating

lymphocyte (TIL) vaccines is: Arias-Pulido et al., 2018, The combined presence of CD20 + B cells and

PD-L1+ tumor-infiltrating lymphocytes in inflammatory breast cancer is prognostic of improved patient

outcome. Breast Cancer Research and Treatment, 2018 June 1. doi: 10.1007/s10549-018-4834-7. In this

paper, the authors measured the levels of proteins PD-L1 (checkpoint inhibitor) and CD20 (tumor marker)

in 221 biopsies of patients with inflammatory breast cancer (IBC) as potential biomarkers of patient

outcomes. Their data showed that the combination of high levels of CD20-positive TILs plus high levels

Page 104: CANCER VACCINES - Worcester Polytechnic Institute

104

of PD-L1-positive TILs correlates with patient disease-free survival. We interviewed the corresponding

author for the paper Anonymous, a cancer vaccine researcher in the Departments of Microbiology and

Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH.

When asked his opinion whether there are mouse xenograft models for inflammatory breast cancer (IBC),

and if so, whether he has tested checkpoint therapy in that model, he responded: “Yes, actually I have a

few IBC [inflammatory breast cancer] cell line models that are being used to evaluate small molecule

inhibitors. And I’m developing humanized PDX [patient-derived xenograft] models, but we have not used

these [PDX] models yet, mainly due to money issues, and the issues of autologous vs. allogeneic immune

cells used for those experiments”. Thus, the cancer vaccine researcher agrees with our assessment that

anti-PD-1 or anti-PD-L1 antibody checkpoint therapy might help supplement his TIL therapy for breast

cancer, and he would like to test the therapy combination in mouse models for breast cancer once money

is not an issue.

When using TIL vaccines, we found that some researchers isolated and expanded in vitro specific

TILs that target a particular tumor antigen, while other researchers attempted to amplify all TILs isolated

from a patient’s tumor (which presumably can target multiple tumor antigens). But is was not clear to us

which approach is best. To shed light on this issue, we interviewed Dr. Emese Zsiros, of the Department

of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, who is a

corresponding author on the paper: Mayor et al., 2018, Adoptive cell transfer using autologous tumor

infiltrating lymphocytes in gynecologic malignancies. Gynecologic Oncology, 2018 May 23. pii: S0090-

8258(18)30920-X. The paper provides an overview of the current clinical results, risks, challenges, and

future applications of TIL-based therapy in gynecologic malignancies. When asked her opinion about

treating gynecologic tumors and whether the best results are obtained by selecting the TILs against a

particular target antigen, or by attempting to amplify all TILs present in a tumor biopsy, she replied: “This

is a tough question. It would be easier to just target one specific universal antigen and engineer cells

against that antigen, but unfortunately we don’t have such a universal antigen expressed by every ovarian

cancer cell in every patient and not expressed by normal cells in the body. There are T cells engineered

against target NY-ESO for example, but only about 20-30% of ovarian cancer patients express that.

Using a diverse group of TILs isolated from tumor tissue is more attractive as they are polyclonal,

however there are many patients where there are essentially no T cells in the tumor microenvironment,

which makes this approach challenging as well. Also resecting tumor tissue from recurrent disease deep

inside the pelvis and or other areas/organs is often challenging and risky in ovarian cancer patients. In an

ideal world, finding a good neoantigen target that is specific would be the best, and just target that”. So,

Dr. Zsiros indicates that each of the two main TIL methods (isolating TILs that target one antigen, or

isolating polyclonal TILs from a resected sample that target multiple antigens) have their advantages and

problems. Using the first method fails to treat tumor cells that lack the target. A universal target does not

exist for ovarian tumors, if it did that would be the way to go. Using the second method with polyclonal

TILs is better as it kills a larger variety of tumor cells, but TILs are not always present and isolatable from

a tumor, and resecting deep ovarian tumors has its problems.

For breast cancer tumors, obtaining tumor samples via core needle biopsy is increasingly

common. But for cancer vaccines, it is unclear whether the biopsy sample is immunologically equal to

the main tumor with respect to TIL cells. One paper that addresses this problem directly is: Cha et al.,

2018, Comparison of tumor-infiltrating lymphocytes of breast cancer in core needle biopsies and resected

specimens: a retrospective analysis. Breast Cancer Research and Treatment, 2018 June 5. doi:

10.1007/s10549-018-4842-7. The authors investigated whether the TIL scores in core needle biopsies

(CNBs) taken from patients with advanced breast cancer are immunologically representative of those

taken from resected specimens. They analyzed 220 matched pairs of CNBs versus their resected

counterparts, scoring stromal TILs on slides stained with H&E. The authors concluded that more than

five CNB cores accurately predicts the TIL score of the entire tumor. We interviewed the first author on

the paper (whose name was forwarded to us from the corresponding author): Dr. Yoon Jin Cha,

Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, 211

Page 105: CANCER VACCINES - Worcester Polytechnic Institute

105

Eonju-ro, Seoul, Republic of Korea. When asked whether she is aware of any study comparing the

anti-tumor therapeutic activity of TILs amplified from core needle biopsies versus TILs prepared from

resected specimens, she responded: “In general, high TILs in breast cancer are known to be a positive

predictive marker of neoadjuvant chemotherapy in HER2 and TNBC tumors. Among the TILs, CD8+ T-

cells may be the most important for the anti-tumor effect, and are known to be elevated in HER2 and

TNBC tumors, which may have association with improved treatment response in those subtypes. With

respect to your question about whether we are aware of any study comparing the anti-tumor therapeutic

activity of TILs amplified from core needle biopsies versus TILs prepared from resected specimens, I am

not aware of any. I think that to compare the anti-tumor activity of TILs in biopsies vs resections, each

subpopulation of TILs (such as CD8, CD4, FOXP3) should be measured via IHC. And the regional

heterogeneity of immune microenvironment should be considered”. So, Dr. Cha indicates that, in general,

high levels of TIL cells present in a breast cancer tumor predict a good prognosis. She also notes that

among the various types of cells present in the TIL population, the presence of high levels of CD8-

positive cells (cytotoxic T-cells) have the most important anti-tumor effect. She was not aware of any

study attempting to determine whether TILs isolated from breast cancer core needle biopsies are as

effective as those isolated from resected specimens, so that can be a future experiment.

Several studies in our review of the literature indicated that therapies with a high number of TIL

cells provides a better prognosis for the patient. A paper related to this topic is: Huang et al., 2018,

Prognostic impact of tumor infiltrating lymphocytes on patients with metastatic urothelial carcinoma

receiving platinum based chemotherapy. Scientific Reports, 2018 May 10; 8(1): 7485. The authors

investigated the prognostic role of TIL levels on patient survival for metastatic urothelial carcinoma

(mUC). The patients also received standard platinum-based chemotherapy. They analyzed 259 mUC

patients, of which 179 (69%) had intense (high) TILs, and 80 (31%) had non-intense (low) TILs. The

median overall survival was 15.7 months for the intense TIL group versus 6.7 months for the non-intense

group (p  < 0.001). The authors conclude that assaying TIL staining intensity (numbers) for mUC patients

is clinically useful for patient risk stratification and counseling. We interviewed the corresponding author

for the paper: Anonymous, Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial

Hospital, Kaohsiung City, Taiwan. When asked whether their data showing that patients with metastatic

urothelial cancer with intense TIL staining have a longer overall survival than those with low intense

staining could be extended to hypothesize that treating mUC patients with high TIL numbers (perfusing a

high number) should be superior than treating with low numbers, he replied: “You are right. The

oncologic outcomes of treating mUC [metastatic urothelial carcinoma] patients with high TIL numbers

are superior than those of treating with low numbers. The immune micro-environment is essential in the

treatment”. Thus, researchers have taken the general finding that high TIL numbers in a tumor correlate

with good patient prognosis, and extended that finding to treat patients with a high number of TILs.

Another paper on the topic of high TIL numbers correlating with good patient prognosis is:

Aghajani et al., 2018, Predictive relevance of programmed cell death protein 1 and tumor-infiltrating

lymphocyte expression in papillary thyroid cancer. Surgery, 2018 Jan; 163(1): 130-136. In this paper, the

authors studied the levels of TIL density and the levels of PD-1 which is produced by some tumor cells

and binds to receptor PD-1 on T-cells to induce a strong inhibition of TILs that have migrated to the

tumor site. The authors investigated the predictive value of assaying PD-1 expression and TIL density in

75 patients with papillary thyroid tumors. Their data showed that PD-1 expression significantly

correlated with increased incidence of lymphovascular invasion (P = .038), extrathyroidal extension

(P = .026), and concurrent lymphocytic thyroiditis (P = .003). In the TIL population, a low presence of

CD8+ and CD3+ (cytotoxic T-cells) correlated with a significantly higher incidence of lymph node

metastasis (P = .042) and extrathyroidal extension (P = .015). A high density of CD8+ TILs was

significantly associated with favorable disease-free survival (P = .017), and the shortest patient survivals

occurred in patients with high PD-1, and low CD8. We interviewed the corresponding author on the

paper: Anonymous, Thyroid Cancer Group, Ingham Institute for Applied Medical Research, Liverpool,

NSW, Australia; and the School of Medicine, Western Sydney University, Campbelltown, NSW,

Page 106: CANCER VACCINES - Worcester Polytechnic Institute

106

Australia. When asked “based on the findings of your paper that patients with papillary thyroid cancer

with high PD-1 expression and low CD8+ cells in the tumors have poor prognosis, we assume it could be

hypothesized that treating such patients with a combination of a high number of TILs (containing CD8+

cells) and anti-PD-1 checkpoint therapy might benefit those patients?” She replied: “It is a possibility,

and there have been some studies investigating the potential of combining PD-1/PD-L1 targeted

immunotherapies with other treatments to increase CD8 T-cell populations. However, the combination

therapy has yet to be investigated in patients with thyroid cancer. So, she agreed that for her papillary

thyroid cancer patients with high PD-1 expression and low TIL CD8+ cells (and very poor prognosis),

those patients might benefit from a combination therapy of a high number of TILs (containing CD8+

cells) and anti-PD-1 checkpoint therapy, although to her knowledge that experiment has not yet been

done.

With respect to the topic of TIL numbers versus patient prognosis, some scientists have

observed a difference between a high number of infiltrating neutrophils versus a high number of CD8+

cytotoxic killer cells. An example is the paper: Liu et al., 2018, The prognostic values of tumor-

infiltrating neutrophils, lymphocytes and neutrophil/lymphocyte rates in bladder urothelial cancer.

Pathology Research and Practice, 2018 May 20. pii: S0344-0338(18)30352-2. In this paper, the authors

investigated the roles of tumor-infiltrating neutrophils (TINs), tumor-infiltrating lymphocytes (TILs), the

neutrophils/lymphocytes ratio (NLR), and clinical outcomes in patients with bladder cancer (BC). They

analyzed 102 bladder cancer patients. Immunohistochemistry was used with CD66b antibodies to score

neutrophils, and with CD8 antibodies to score lymphocytes. Their results indicated that high TINs and

high NLR ratios associated with poor overall patient survival, while higher TILs correlated with longer

survivals (P < 0.01). We interviewed the corresponding author on the article: Dr. Erlin Sun, Department

of Urology, Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China.

When asked his opinion of why the high tumor-infiltrating neutrophil count correlated with poor patient

outcome, while high TILs correlate with better patient prognosis, he replied: “We know that neutrophils

can be polarized into either an anti-tumoral (N1) or a pro-tumoral (N2) phenotype, so they show different

functions. N1 neutrophils are anti-tumoral effector cells by inducing cytotoxicity, mediating tumor

rejection and anti-tumoral immune memory. In contrast, N2 phenotype neutrophils support tumor

progression by promoting angiogenesis, invasion, metastasis and immunosuppression. According to the

results of our study, we suspect that tumor-infiltrating neutrophils have pro-carcinogenic [N2 phenotype]

effects on tumor progression. However, tumor-infiltrating CD8+ lymphocytes are found to be favorable

prognostic factors in our study, which may play a role in tumor suppression by immune process. Our

results are just a clinical retrospective analysis, the mechanism still need to be researched”. So, Dr. Sun

thinks that the reason a high tumor-infiltrating neutrophil count correlates with poor patient prognosis is

that the infiltrating neutrophils are of a N2 phenotype that is pro-tumorigenic. N2 neutrophils promote

angiogenesis, invasion, metastasis, and immunosuppression. So, it will be important in future studies to

assay for exactly which types of immune cells have migrated to the tumor site.

Results for CAR Vaccines A problem noted throughout our Lit Review is the induction of side-effects caused by the

vaccine. In the case of CAR vaccines, the side-effects can be caused by the presence of target antigen in

normal tissues (so the T-cells target and kill those normal cells). An example of this is the paper:

Richman et al., 2018, High-Affinity GD2-Specific CAR T Cells Induce Fatal Encephalitis in a Preclinical

Neuroblastoma Model, Cancer Immunology Research, 2018 Jan; 6(1): 36-46. In this article, the authors

studied neuroblastoma xenograft mouse models with a vaccine containing GD2-CAR T-cells. The CARs

showed enhanced anti-tumor activity, but in some cases the treatment caused death of the mice. The

authors observed brain T-cell infiltration and proliferation (which seems necessary for killing the tumor

cells), but the T-cells may have also targeted nearby GD2 in normal brain cells. We interviewed the

Page 107: CANCER VACCINES - Worcester Polytechnic Institute

107

corresponding author for the paper: Dr. Michael C. Milone, Center for Cellular Immunotherapies,

Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania. When asked

his opinion whether targeting GD2 in normal cells can be avoided in human patients, he replied: “…The

T cell activity in the brain is due to targeting low level GD2 expression on neurons and glial cells. While

some scientists believe it is possible to design a CAR T-cell therapy that can distinguish the generally

higher expression in tumors compared to the normal expression, I think this will be challenging with

current technology”. So, Dr. Milone agrees that the sometimes fatal encephalitis he observes in his mouse

models treated with GD2-CAR cells likely results from the targeting of GD2 in normal neurons and glial

cells expressing low levels of GD2. He believes it will be challenging to design a CAR to distinguish the

high GD2 expressors from the low expressors. So, more research should be done identifying antigens

almost exclusively expressed in the tumors, if possible.

Another example paper dealing with CAR-induced side-effects is Locke et al., 2017, Phase 1

Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory

Aggressive Lymphoma. Molecular Therapy, 2017 Jan 4; 25(1): 285-295. In this study, the authors

designed CAR KTE-C19, an autologous CAR T-cell therapy targeting CD19 that also uses CD3-zeta and

CD28 as co-stimulators to treat patients with refractory B-cell leukemia. The CAR was was used to treat

7 patients. 1/7 patients (14%) experienced dose-limiting toxicity of grade-4 cytokine release syndrome

(CRS), 1/7 patients (14%) showed grade >3 CRS, and 4/7 patients (57%) showed neurotoxicity. All

>grade-3 events resolved within 1 month. 3/7 patients showed clinical responses (stabilizations) at 12

months. To get an idea of the main cause of the side-effects, we interviewed the corresponding author for

the paper: Dr. Frederick L. Locke, MD, Department of Blood and Marrow Transplantation, Moffitt

Cancer Center, Tampa, FL. When asked his opinion on the cause of the neurotoxicity observed in 4 of

your 7 (57%) phase-I patients, he stated: “More and more evidence suggests that there is breakdown of

the blood brain barrier, so the CAR T-cells get into the CSF where they continue to secrete cytokines.

This likely leads to endothelial damage and dysfunction, which appears to be the main driver of CNS

toxicity”. So, Dr. Locke indicates that the evidence from his lab suggests the neurotoxicity observed with

his KTE-C19 CAR therapy is caused by a breakdown of the blood brain barrier (BBB), allowing CAR

cells to enter the cerebrospinal fluid (CSF), and in a deteriorating cycle of events the CAR T-cells

continue to secrete cytokines in the CSF that causes endothelial cell damage. Thus, the side-effects are not

only caused by a targeting of a specific antigen in normal cells, it is also caused by the continued elevated

production of cytokines by the T-cells.

The most frequently observed side-effect is cytokine release syndrome (CRS). Thankfully, in

most cases, the CRS was manageable by treating with corticosteroids and IL-6-receptor blocking

antibodies. In order to shed light on how cancer vaccine treatments can cause CRS, we interviewed the

corresponding author on a paper where CRS was observed: Young et al., 2018, Activity of Anti-CD19

Chimeric Antigen Receptor T Cells Against B Cell Lymphoma Is Enhanced by Antibody-Targeted

Interferon-Alpha. Journal of Interferon and Cytokine Research, 2018 Jun; 38(6): 239-254. The

corresponding author is: Dr. John M. Timmerman, Division of Hematology & Oncology, Department

of Medicine, Center for Health Sciences, University of California at Los Angeles, Los Angeles, CA. In

this paper, the authors investigated methods for lengthening the survival of CD19-CARs in patients with

B-cell lymphomas. Because interferons (IFNs) have the ability to promote T-cell activation and survival,

the authors tested whether antibody-targeted IFN therapy could enhance their CAR therapy. They

produced a new type of CAR anti-CD20-IFN, containing an antibody-like portion against the CD20

lymphoma target fused with a potent type-1 IFN isoform alpha14 (α14). The combination approach was

found to enhance lymphoma cell killing in vitro. Pre-treatment of the lymphoma cell lines with the fusion

peptide (anti-CD20-hIFNα14) markedly increased cytokine production by the subsequently added CARs,

enhancing several CAR activities, but it was unclear to us whether this novel approach might increase the

incidence of cytokine release syndrome. When asked whether his new anti-CD20-hIFNα14 treatment

might cause increased cytokine release syndrome, Dr. Timmerman replied: “Maybe in some patients. But

there are many patients that don't have a clinical response [to the CD19-CAR] or much CRS, so if we

Page 108: CANCER VACCINES - Worcester Polytechnic Institute

108

knew how to predict those patients, adding the [anti-CD20-hIFNα14] fusion protein might help them”.

So, Dr. Timmerman agreed with us that his new drug could indeed increase the incidence of cytokine

release syndrome, but that the drug is strongly needed to treat the lymphoma patients that do not respond

to CD19-CARs, as it targets a different antigen (CD20) on the lymphoma cells, so this warrants its use.

Sometimes tumor location is important. For example, solid tumors are difficult to treat with

cancer vaccines due to their location and general inaccessibility. Over the past decade, the use of CAR

cells has led to strong improvements in patients with hematopoietic malignancies which are readily

treated by systemic infusions on of the CAR cells. But CAR treatment of solid tumors is hindered by

challenges inherent to an organized tumor mass, such as abnormal vasculature, migration through a dense

stroma, and an elevated tumor interstitial pressure (IFP). Some scientists are trying to overcome these

problems by performing a localized delivery of the CARs directly to the tumor. An example of this is the

paper: Hardaway et al., 2018, Regional Infusion of Chimeric Antigen Receptor T Cells to Overcome

Barriers for Solid Tumor Immunotherapy. Journal of Vascular and Interventional Radiology, 2018 Jul;

29(7): 1017-1021. The authors take advantage of the fact that hepatic tumors derive their blood supply

from the hepatic arterial circulation (where the scientists deliver CARs), while normal hepatocytes mainly

subsist off the portal circulation (not encountered by the CARs). This regional delivery approach has

already been used with chemotherapeutic, radiotherapeutic, and chemoembolic agents for liver

tumors. The regional approach has also been used for the treatment of of hepatic and pancreatic

malignancies with dendritic cells, macrophages, or lymphokine-activated killer cells (LAKs). We

interviewed the corresponding author for the paper: Dr. Steven C. Katz, MD, FACS, Associate

Professor of Surgery, Director, Complex General Surgical Oncology Fellowship, Director, Office of

Therapeutic Development, Roger Williams Medical Center, Providence, Rhode Island. When asked his

opinion about whether his regional CAR delivery method would not only treat the cancer but help

diminish the adverse side-effects, he replied: “We believe the overall therapeutic index would be

enhanced, in general, for regionally infused products”. So, Dr. Katz agreed that his regional method for

delivering CAR T-cells to solid tumors not only reduces the tumors better, it also decreases the adverse

side-effects.

With respect to CAR vaccines, some researchers are working on new methods for delivering the

CAR receptor gene to the T-cells without using lentiviruses (non-lentiviral delivery methods). The

lentiviruses tend to integrate at random sites and can be harmful. A new technique delivers the RNA

encoding the CAR receptor directly to the T-cells, as seen in the paper: Svoboda et al., 2018, Non-viral

RNA chimeric antigen receptor modified T cells in patients with Hodgkin lymphoma. Blood, 2018 Jun

20. pii: blood-2018-03-837609. The authors treated 4-5 patients with classical Hodgkin lymphoma (cHL)

using CAR T-cells. To limit potential toxicities, they used non-viral RNA CART19 cells, which the

authors expected to express the CAR receptor protein only for a few days off the delivered RNA, as

opposed to CARs generated by viral vector transduction which expand in vivo and retain CAR expression

via the integrated DNA. Their results showed no severe toxicities. To our knowledge, this is the first

CART19 clinical trial to use non-viral RNA gene delivery. But it was unclear to us how efficient the

RNA technique was relative to the highly efficient lentivirus technique, so we interviewed the

corresponding author on the paper: Dr. Jakub Svoboda, Lymphoma Program, Abramson Cancer Center,

University of Pennsylvania, Philadelphia, PA. When asked what percent of the patient’s T-cells can be

treated with the RNA technique versus treatment with a lentivirus encoding the CAR gene, he replied:

“That is a good question, but difficult to answer since every patient is different. [In our study] the number

of infused modified RNA CART19 cells varied (the dose was based on weight, the protocol allowed for a

range). These cells [RNA-treated CAR cells] are transient (last few days, do not expand) and they likely

represent only a tiny portion of the lymphocytes in relation to the total number in the human body (the

total number of lymphocytes in the human body is estimated to be about 2x1012). For the lentivirus

transduced CART19 cells, these tend to expand in vivo, but the expansion varies from patient to patient,

but again - likely a tiny portion”. So, Dr. Svoboda indicated that it is difficult to determine the percent of

T-cells transduced with his RNA technique to deliver the CAR gene versus using a lentivirus, but in any

Page 109: CANCER VACCINES - Worcester Polytechnic Institute

109

case, the end numbers vary for each patient depending on the extent of CAR expansion, not just based on

the percent transfected.

Results for Checkpoint Inhibitor Treatments

One of the recent trends in the checkpoint vaccine field is uncovering various ways that some

tumors are resistant to checkpoint therapy. For example, an exciting find in 2018 was the discovery

that a high level of TGFβ hormone in the tumor micro-environment acts as a primary mechanism of

immune evasion for tumor cells (Tauriello et al., TGFβ drives immune evasion in genetically

reconstituted colon cancer metastasis, Nature, 2018 Feb 22; 554: 538-568). In order to gain information

on the feasibility of using a combination of a checkpoint therapy with anti-TGFβ antibody, and

determining how TGFβ levels might be assayed in patients, we interviewed the first author on the article

(whose name was provided by the corresponding author): Dr. Daniele Tauriello, Institute for Research in

Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.

When asked her opinion of whether a clinical trial might be feasible combining a checkpoint inhibitor

vaccine with anti-TGFβ antibody, she replied: “We think that, at least in CRCs [colon cancer

metastases], high levels of TGFβ prevent or strongly diminish a full activation of T cells. Furthermore, T

cell infiltration is low, which could be a direct result of T cell dysfunction, but might also be a separate

mechanism by which TGFβ affects immune evasion. If these are the two main ways in which high TGFβ

levels suppress anti-tumour immunity, then inhibition of the TGFβ pathway (for example with TGFBRI

inhibitor Galunisertib), should lead to full activation of T cells….. and a combined inhibition of stromal

TGFβ signalling with blockade of the PD-1/PD-L1 checkpoint could be an effective way to overcome

immune evasion in patients with advanced/metastatic CRC”. When further asked whether there is an

assay for clinicians to quantitate the levels of TGFβ in the tumor microenvironment (to predict which

patients might benefit from the combined therapy), she responded: “….in our lab's previous papers we

showed that high TGFβ mRNA levels strongly predict poor prognosis in stage I+II+III patients, so if

primary tumours could be removed by surgeons and analysed for TGFβ mRNA levels (or analyzed for

TGFβ-controlled gene expression levels), …we expect that the combined insight will lead to more power

to clinically intervene”. So, Dr. Tauriello indicated that the way to measure TGFβ levels in patients

would be for the surgeons to remove the primary tumors, and then for lab personnel to assay for for TGF-

beta gene transcription levels (presumably using something like RT-PCR). The high TGFβ patients would

have a poorer prognosis but should respond to their dual therapy.

All cancer immuno-therapies have side-effects, and checkpoint inhibitor treatments are no

exception. But the presence of side-effects does not necessarily mean the drugs should be discontinued.

In most cases, the side-effects observed were mild and transient, and are likely far outweighed by the poor

prognosis of a relapsing cancer patient. To gain more insight on the topic of checkpoint inhibitor side-

effects, we interviewed an expert on liver injuries induced by checkpoint inhibitor treatments, Dr.

Eleonora DeMartin, Centre Hépatobiliaire, Hôpital Paul Brousse, Groupe Hospitalier Paris Sud, DHU

Hepatinov, Villejuif, France. Dr. DeMartin’s contact information was provided by the corresponding

author of the paper: De Martin et al., 2018, Characterization of liver injury induced by cancer

immunotherapy using immune checkpoint inhibitors, Journal of Hepatology, 2018 Jun; 68(6): 1181-1190.

When asked her opinion of whether the benefits of checkpoint inhibitor therapy outweigh the side-effects

caused by the treatments, she replied: “I agree that most of the immune-mediated hepatitis side-effects

provide a far better prognosis than the prognosis from cancer relapse/progression. Liver injury induced by

cancer immunotherapy either improves spontaneously, or it responds to corticosteroid therapy in most of

the patients. Interestingly, it seems that patients who develop immune-related adverse-effects (iRAEs)

better respond to therapy. However this finding needs to be confirmed”. When asked whether there is any

way for a clinician to predict which patients are more likely to develop liver injuries following checkpoint

therapy, she replied: “No, unfortunately we haven't been able to identify predictive factors for hepatic

Page 110: CANCER VACCINES - Worcester Polytechnic Institute

110

IRAEs. We are running a multidisciplinary study now in order to answer this question”. When asked

whether she thinks that autoimmune diseases resulting from checkpoint inhibitors are severe enough and

common enough to prohibit inhibitor usage, she replied: “The answer is no. They remain a rare

complication, and in most cases they are not severe. Considering the revolutionary results of cancer

immunotherapy I encourage its use”. So, Dr. DeMartin, indicated that in the case of her patients treated

with checkpoint therapies, the side-effects are minor or rare, and are less important than attempting to

save the patient’s life from a relapsing cancer.

Continuing on the topic of side-effects caused by checkpoint therapies, we interviewed Dr.

Michael A. Postow, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New

York. Dr. Postow was corresponding author on: Postow MA, Sidlow R, Hellmann MD (2018) Immune-

Related Adverse Events Associated with Immune Checkpoint Blockade. New England Journal of

Medicine, 2018 Jan 11; 378(2): 158-168. When asked his opinion about whether the side-effects are

outweighed by the potential benefits of the checkpoint therapy, he replied: “I think the benefits

dramatically outweigh the possibility of side-effects. Untreated metastatic cancer invariably leads to

death at some point. Immunotherapies offer patients the hope that they can keep their cancer under

control for a long time if they respond well. This is an amazing potential benefit and well worth the

possibility of side-effects. And even if side-effects occur, they can be well managed and do not usually

lead to permanent disability. The side-effects usually resolve within weeks to months”. When asked

whether there is any way for physician’s to determine which patients will develop side-effects, he

responded: “Unfortunately we do not really know. There are many research programs going on to

identify predictive factors that are associated with immunotherapy side-effects. It may have to do with

germline genetics, or with other “host” immunologic factors. Some people think that patients with less

heavy of a burden of cancer are more likely to have side effects, perhaps because these patients with

lower tumor volume are less immunosuppressed”. And when asked his opinion about whether there is a

subset of patients that should absolutely not receive checkpoint therapy, such as immunocompromised

patients or patients on immunosuppressant drugs, he replied” Although efficacy may be a little lower in

patients on immunosuppressants or otherwise immunocompromised, since these patients have been

shown to benefit from immunotherapy in some situations, I think it is reasonable to try immunotherapy

when no other good cancer treatments are available”. Thus, Dr. Postow is a strong advocate of attempting

to use immunotherapies on patients with otherwise very poor prognoses, even if some side-effects

develop.

One serious side effect of checkpoint inhibitor therapy is the occasional development of T-cell

tumors. The point of checkpoint therapy is to activate the patient’s inhibited T-cells, and sometimes

tumors develop. A paper covering this topic is: Ludin and Zon, 2017, Cancer immunotherapy: The dark

side of PD-1 receptor inhibition. Nature, 2017 Dec 7; 552(7683): 41-42. doi: 10.1038/nature24759.

We interviewed Dr. Aya Ludin Tal, Postdoctoral Fellow, Zon Lab, Harvard Department of Stem Cell

and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA. Dr. Tal’s

contact information was provided by Dr. Zon, the corresponding author on the paper. When asked

whether there is any way that physician’s can determine which patients will develop T-cell tumors, she

replied: “…. In some cases, the patient already has a T-cell tumor, [and in these cases] various options of

treatment should be considered. ….If their T cells are cancerous, it might not be a good idea to expand

this population of cancer cells by driving them into proliferation using PD-1. PD-1 inhibition is not yet

used in hospitals with patients with T cell-based cancers, but since it is gaining interest in multiple types

of cancers, the authors warn against possible side effect that is not seen in other types of cancer”. Their

paper also showed that anti-PD-1 activates different subsets of T-cells, so we asked her which types are

more effective for their patients. She replied: “Different subsets of T cells have different roles in fighting

or protecting cancer. Cytotoxic [CD8+] and helper cells [CD4+] help fight tumors, whereas regulatory T

cells [T-reg] can protect the tumors”. And when asked whether the overall risks of immunotherapy are

different or on par with other cancer treatments like radiation or surgery, she replied: Each treatment has

its own risks, so it’s hard to say. It also depends on the type of immunotherapy used, each has its own

Page 111: CANCER VACCINES - Worcester Polytechnic Institute

111

risk, such as the induction of autoimmune effects through CAR T-cell administration (uncontrolled T

cells). The risks of the different immunotherapies are still being investigated. However, anti PD-1 and

anti-CTLA-4 therapies have already been approved clinically for many types of cancers, suggesting that

the benefit for the patient is higher than the risk. The efficiency of treatment, whether it be

immunotherapy, surgery or irradiation is an important factor considered, and many times these treatments

are combined together”. Thus Dr. Tal indicated that checkpoint therapy is not yet used clinically for

patients with T-cell tumors (as it could further activate the tumors), but warns the current emphasis on

expanding checkpoint therapy to other types of cancers should consider the effects on T-cells. And with

respect to the overall risk, she pointed out that checkpoint therapies have already received FDA approval,

so a panel of experts have already determined that the benefits outweigh the risks.

An interesting recent finding in the checkpoint inhibitor field is the discovery that the gut

microbiome (the type and quantity of bacteria present in the gut) strongly affects the response to

checkpoint therapy. An example is the article: Gopalakrishnan et al., 2018, Gut microbiome modulates

response to anti-PD-1 immunotherapy in melanoma patients, Science, 2018 Jan 5: 359(6371): 97-103.

We interviewed the first author on this paper Dr. Vancheswaran Gopalakrishnan, Department of

Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.

Dr. Gopalakrishnan’s contact information was provided by the corresponding author on the paper. One of

the findings of the paper was that patients with a gut microbiome enriched for Ruminococcaceae family

bacteria respond better to anti-PD-1 therapy, so we asked Dr. Goplakrishnan whether they plan to move

forward with clinical trials by supplementing patient microbiomes with Ruminococcaceae microbes? He

replied: “We are planning a clinical trial to test the hypothesis that modulation of the gut microbiome will

enhance therapeutic responses. Several strategies will be used, including fecal microbiota transplant and

designer probiotics that enrich for favorable bacteria……. FMT [fecal microbiota transplants] have

already proven to be successful for treatment of Clostridium difficile infections, but there is limited data

for oncology [and FMTs]. In addition to the above, one may also consider lifestyle changes such as

dietary modifications to modulate/maintain the microbiome”. When asked how difficult it is to assay a

patient’s microbiome, he replied: “This can be done in several ways. The most commonly used approach

is called 16S rRNA sequencing. The 16S gene is a ubiquitous bacterial marker that has both conserved

and variable regions. The conserved regions act as targets for PCR-primers. The variable regions can be

amplified, sequenced and compared with reference databases to identify bacterial diversity and

composition. Another approach is whole genome shotgun sequencing. This gives greater resolution of

bacteria at the species level and also gives a sense of their functional capabilities. But sequencing takes

time, with a turnaround of approximately 1 month, so this is not yet feasible at a per-clinic basis. But this

is surely something that people are invested in”. Thus, Dr. Gopalakrishnan provided some very useful

information on: 1) how indeed they are moving forward to test their hypothesis that modifying the gut

microbiome affects the success of PD-1 therapy, 2) there are several different ways they plan to modify

the microbiome in patients (including fecal transplants and probiotics), and 3) the methods for assaying

species type and number in the biome include sequencing ribosomal RNAs, and performing

bioinformatics to determine the species type and abundance.

Page 112: CANCER VACCINES - Worcester Polytechnic Institute

112

CONCLUSIONS and RECOMMENDATIONS

Based on the research performed for this IQP project, our team has developed several conclusions

and recommendations. We focused on two main problems prevalent in the area of cancer vaccines: 1) the

fact that only a subset of patients respond to a particular therapy, and 2) the fact that most cancer vaccines

cause adverse side-effects. We emphasized new advances in the field for overcoming these problems.

Cancer Vaccine Non-Responding Patients

With respect to the topic of non-responding patients, our research indicates that tumors resistant

to cancer vaccine treatments occur for all types of cancer vaccines and for all types of cancers. These

tumors are non-responsive for a variety of reasons. Some of the most important reasons are discussed

below along with potential solutions:

1) Down-Regulation of the Target Antigen: The major mode of resistance of some tumors to immune

therapy is a loss of target antigen on the surface of the tumor cells. For reasons that are not yet

understood, in some cases a patient’s tumor stops making the target antigen, or lowers its expression.

When this occurs, a potential remedy is to follow the initial failed therapy with a different one that targets

another antigen, if available. This combination approach is most feasible for the lymphomas and

leukemia where multiple target antigens are already available. One of the best practices we observed in

our review of the cancer vaccine clinical trials was the constant monitoring of target antigen expression

on the tumor. This is easily done by using immunohistochemistry (IHC) to measure the levels of target

protein, or by using reverse transcriptase polymerase chain reaction (RT-PCR) to measure the levels of

target mRNAs. The best patient prognoses were observed for patients with tumors strongly expressing

the target antigen.

2) Tumor Heterogeneity: A recent discovery in the cancer field is that some tumors are not

homogeneous; they consist of regions that differ from each other. In some cases, portions of the tumor

may lack expression of the target antigen, so those cells will not be killed by the vaccine and will continue

growing. In other cases, different areas of a tumor express different neo-antigens (new proteins made by

a tumor caused by different DNA mutations over time). In these cases, different areas of the tumor are

genetically distinct from each other. Each has their own growth rates, metabolic pathways, and overall

aggression. In these cases, if it is possible to resect genetically different parts of the tumor without

harming the patient, perhaps different tumor regions can be identified with their corresponding antigens,

then the patient could be treated with a combination vaccine against the antigens of each section.

3) Immunosuppression Induced by the Tumor: In some cases, the patient’s tumor inactivates the

patient’s immune system, lowering removal of a cancer (or blocking a cancer vaccine). In some cases,

this inactivation occurs via checkpoint inhibition on nearby T-cells to keep them in check. This immuno-

suppression keeps the T-cell from killing the tumor. The discovery of key components of checkpoint

inhibition (including PD-1, PD-L1, CTLA-4) has in recent years allowed the use of antibodies against the

components to re-activate the patient’s immune system. This type of cancer “vaccine” has increased

exponentially in the past few years. A very recent 2018 discovery is the strong role of TGFβ in the tumor

microenvironment at inhibiting T-cells or DCs that have migrated to the tumor, and this finding opens up

the possibility of treating these resistant patients with anti-TGFβ antibodies, or with antibodies that block

TGFβ signaling.

4) Immunocompromised Patients: In some cases the patient is immunocompromised due to their pre-

treatment with chemotherapy (which destroys actively dividing cells in the body). In these cases, the

patient may lack the necessary immune components for killing the tumor. In the case of monovalent

Page 113: CANCER VACCINES - Worcester Polytechnic Institute

113

antibody treatments, the binding of the vaccine antibody to the tumor cell by itself does not kill the cell,

and other components of the immune system must recognize the bound antibody and help kill the cell. In

the case of bispecific antibodies designed to bind the target antigen on the tumor cell while also binding to

a T-cell to bring them together, an immunocompromised patient may lack a sufficient number of T-cells

to be recruited to the site. So, perhaps these patients could be treated with an ADC vaccine or a CAR T-

cell vaccine, which by themselves can kill a tumor cell.

5) Short Vaccine Half-Life: In some cases, the cancer vaccine was ineffective in a patient due to a short

half-life. This was especially a problem with passively administered antibody vaccines that are rapidly

cleared from the body. A recent trend with antibody vaccines is to encode the antibody genes in a

deliverable vector (such as a harmless virus), and deliver the vector to the patient’s bloodstream. The

DNA continues to express the antibody gene far longer than with a delivered protein antibody. The short

half-life of some CAR T-cell vaccines appears to have resulted from a lack of co-activation of the T-cells,

but this problem has now been remedied through the inclusion of co-stimulation domains on the

engineered CAR receptor (such as CD28 or CD3-zeta).

6) Altered Tumor Growth Pathways: In some cases, a tumor became resistant to therapy when the

tumor mutated to switch growth pathways. Thus, the original vaccine treatment attempting to block the

pathway failed. In these cases, perhaps the tumor resistance could be overcome by switching drugs to

block the new growth signaling pathway the tumor now depends on. This is especially a problem when

using antibody vaccines to block specific receptors. If the tumor is no longer dependent on that particular

signaling pathway, blocking the pathway’s receptor will not prevent tumor growth.

Cancer Vaccine Side-Effects

With respect to the topic of side-effects, our research shows that all types of cancer vaccines

cause adverse side-effects. The side-effects varied considerably, depending on the type of treatment and

the type of cancer, ranging from very mild and transient, to patient death in a few cases.

1) Patient Deaths: In clinical trial, three patients died from E. coli or Candida sepsis caused by treatment

with a CD19 x CD3 bispecific antibody that eliminated B-cells from the leukemia patients, hindering their

ability to make antibodies against the endogenous pathogens. And seven patients died in one year (2016)

in CAR clinical trials when the CAR cells caused fatal brain swelling. So, patient deaths indeed

occasionally occur with cancer vaccines, and these deaths are not unexpected for patients with relapsed

cancers. However, the patient deaths are rare from the vaccines, and most of our interviewees argued are

worth the cost of trying to save lives from certainly fatal relapsing cancers.

2) Cytokine-Release Syndrome: The second most serious side-effect seen with cancer vaccines is

cytokine release syndrome (CRS). CRS is a hyper-elevation of cytokine hormones that typically cause

fevers, hypotension, and hypoxia, but can be deadly when it causes organ failure. Although the cytokine

elevation is actually a desired outcome of cancer vaccines as an outcome of immune system activation,

the activation can become too prolonged and harm the patient. CRS was sometimes seen in the antibody

therapy trials, and in the adoptive T-cell therapies. In a large trial of patients with aggressive non-

Hodgkin’s lymphoma, about 18% of the patients developed CRS. But in most cases, the CRS was

treatable with corticosteroids (to lower inflammation) and with cytokine blocking antibodies (such as

anti-IL6-receptor antibody), so it was not usually life threatening.

3) Autoimmune Disorders: In the case of checkpoint vaccines, which are designed to ramp up the

patient’s immune system, one of the more serious side-effects observed was autoimmune disease (where

the patient’s immune system attacks the body). This is especially a problem with autoimmune

myocarditis, an immune attack on the heart, which can be fatal. While we identified a few papers that

Page 114: CANCER VACCINES - Worcester Polytechnic Institute

114

warned of such events, our interviews with physicians performing these therapies indicated they saw

these disorders only rarely in their patients.

Scientists are still trying to determine what causes the side-effects, but they appear to be caused

by a variety of mechanisms. In some cases, they appear to be caused by expression of the target antigen

in normal cells, thus those cells are destroyed by the therapy. Our interviews with scientists who designed

vaccines showed that designing a cancer vaccine that can only target a tumor cell expressing high antigen

levels while ignoring normal cells with medium expression levels of the antigen is not currently possible.

So, we recommend continuing the search for new target antigens that are only found in tumor cells and

not in normal cells.

In other cases, the killing of normal cells in the body appears to have resulted from a “bystander

effect”. In this case, the normal cells are killed due to their location near a targeted cell. This is

especially the case for antibody-drug conjugate vaccines (ADCs) where the highly cytotoxic drug can be

released nearby normal cells. While this killing could be beneficial if the bystander cell is a tumor cell no

longer expressing target antigen, in most cases the bystander effect is against normal tissue.

Overall, the side-effects in the vast majority of cases were relatively mild grade-1 and -2 events,

transient (lasting only a few days), and treatable. The side-effects were especially treatable if they were

detected early, so these events should be constantly monitored throughout the entire treatment. With

respect to whether the chance of side-effects should deter physicians from using these treatments, the

individuals interviewed for our project were strongly in favor of continuing the treatments, arguing that

the prognosis from the side-effects was far better than very poor prognosis they patients face from the

relapsing cancer. The side-effects they observed were relatively minor or rare, and are less important than

attempting to save the patient’s life from their relapsing cancer. As stated previously, most of the patients

participating in these cancer vaccine trials have run out of other treatment options.

Future Trends

We have observed several trends in the cancer vaccine field that hopefully will help solve some

of the problems mentioned above.

1) Combination Therapies. The simultaneous or consecutive use of two types of therapies has increased

drastically in the past few years. In some cases, the physicians are attempting to treat patients that have

down-regulated one target antigen by using an agent that targets a different antigen. In other cases, the

combination approach is designed to kill the tumor by two different mechanisms which hopefully will

synergize. A good example of this is the combined use of a checkpoint vaccine (to remove the immuno-

suppression caused by the tumor) with a second vaccine to target the tumor (such as a DC, TIL, or CAR).

2) Altered Method of Cell Killing: In some cases, tumor remission was achieved by switching from

recruiting T-cells to the tumor, to recruiting natural killer (NK) cells to do the killing. This has especially

been effective with bispecific antibodies, where one domain is against CD16A on NKs (to recruit them)

while the other domain is against CD30 (targeting the NKs to the lymphoma cells). The approach was

successful and is now being tried in other tumors.

3) Careful Patient Selection: Due to some of the mechanisms discussed above for tumors blocking the

immune system, a recent strategy is to closely monitor the patient’s tumor for potential upregulation of

inhibitors such as TGFβ, PD-1, PD-L1, or CTLA-4. When any of these are found to be elevated in the

tumor microenvironment, the appropriate modifier should be delivered, such as delivering antibodies

against TGFβ signaling if TGFβ is found to be elevated.

Page 115: CANCER VACCINES - Worcester Polytechnic Institute

115

4) Personalized Medicine: Neo-antigens are newly expressed proteins (or carbohydrates) on the surface

of tumor cells that form following DNA mutations. Because neo-antigens are not expressed during

human development, they are viewed as foreign in the body once they are expressed, so they make good

target antigens. The use of new rapid DNA sequencing methods allows a patient’s tumor cells to be

sequenced to analyze for neo-antigen formation, and if found, these could be used to prime DC or TIL

vaccines. But the process needs to be fast, as some patients have a very poor prognosis, and can die

within weeks.

5) CRISPR/Cas9 Editing of CAR T-Cells: One of the most promising and recent innovative uses of

CAR cells combines CAR receptor engineering with the use of CRISPR/cas9 gene editing technology to

eliminate a host gene. For example, the CRISPR system has been used to eliminate the gene TRAC

which is associated with allo-recognition in the body. Thus, its elimination could result in “universal”

CAR cells that could be used in any patient without rejection, eliminating the need to isolate the T-cells

from each patient.

6) Gut Microbiome: An interesting recent finding in the checkpoint inhibitor field is the discovery that

the gut microbiome, the type and quantity of bacteria present in the gut, strongly affects the response to

immune checkpoint therapy. The microbiota from mice non-responsive to checkpoint therapy when

transplanted into naïve mice induced them to become non-responders, and microbiota from responder

mice when transplanted into naïve mice made them become responders. One research team found that

mice with a gut microbiome enriched for Ruminococcaceae type bacteria respond better to checkpoint

therapy. Our interviews with the lead scientist of this study indicated he plans on moving into clinical

trials, either by supplementing the patient’s diet with this type of bacterium, or by using fecal microbiota

transplants (FMTs) enriched for this bacterium.

7) TIL Numbers and Composition: Several studies have shown that the presence of a high number of

tumor-infiltrating lymphocytes (TILs) in a patient’s tumor correlates with a better patient prognosis. This

finding has now been extended to show that treating patients with a high number of TILs is more effective

than using low numbers. In addition, we learned from our interviews that it is important to determine the

composition of the patient’s TILs, because a high number of infiltrating CD8+ (cytotoxic T-lymphocytes)

correlates with a good prognosis, while a high number of infiltrating neutrophils correlates with a poor

prognosis. It was recently shown that infiltrating neutrophils are of an N2 phenotype which is pro-

tumorigenic (promoting tumor angiogenesis, invasion, metastasis, and immunosuppression). So, it will be

important in future studies to assay for exactly which types of cells have migrated to the tumor site.

8) Tumor Location: Diffuse tumors such as lymphomas and leukemia have been treated by the

intravenous perfusion of cancer vaccines for several years now. But solid tumors are difficult to treat due

to challenges inherent to an organized tumor mass, such as abnormal vasculature, migration through a

dense stroma, and an elevated tumor interstitial pressure (IFP). It was recently shown that some of these

problems can be overcome by using a localized delivery of the vaccine directly into (or nearby) the solid

tumor. Our interviews indicated this localized delivery approach might also lower the incidence of side-

effects, as fewer tissues in the body would encounter the vaccine.

Overall, we conclude that cancer vaccines represent a fascinating method for fighting cancer, and

in some cases these approaches have provided complete remissions for relapsing cancers that are

impossible to treat using any other approach. The cancer vaccine field is complex (with treatments

ranging from simple antibody treatments, to genetically modifying T-cells), but the field provides a

variety of approaches for potentially treating a patient’s tumor. While each type of therapy can induce

side-effects in a portion of the patients, we agree with our interviewees that the side-effects are usually

minor, transient, and treatable, and are far less important than attempting to save the patient’s life from

their relapsing certainly fatal cancer.

Page 116: CANCER VACCINES - Worcester Polytechnic Institute

116

APPENDIX

SAMPLE QUESTIONS 1. Cancer Vaccine Side-Effects:

A. Do you agree that most cancer vaccines cause side-effects?

B. In your clinical trials have you observed any serious (grade-3 and grade-4) side-effects?

C. If the side-effects were serious, were they at least treatable (manageable)?

D. If the side-effects were manageable, does the potential for a cure for a patient with a fatal

disease outweigh the risk associated with a potentially fatal side-effect?

E. In your opinion, how can cancer vaccines be improved to cause fewer side-effects? Do we

need more research to identify target antigens on different types of cancers that are not

present in normal cells? Would a personalized medicine approach to identify patient-

specific neo-antigens on a patient’s tumor help decrease off target side-effects?

2. Lack of Vaccine Efficacy:

A. In some cases, a cancer vaccine appears to not work in a particular patient. In your opinion,

what kinds of events can cause this: loss of expression of the target antigen? Inactivation

of the perfused T-cells by the patient’s tumor cells? Lack of vaccine accessibility to a

solid tumor site?

B. For patients who do NOT respond to a cancer vaccine treatment, do you think it would be

beneficial to test a combination treatment with two or more vaccines that work by

different mechanisms?

3. Best Correlates of Protection:

A. It is not clear to us what factors best correlate with a vaccine’s success. For TIL and CAR T-

cell vaccines, some studies indicate success correlates best with a high load of TIL or

CAR cells. This suggests that developing methods for amplifying the number of T-cells

perfused into the patient should be a high priority. Other studies show the worst side-

effects occur at the highest T-cell doses. What is your opinion?

B. TIL vaccines isolated from a patient sometimes contain a mix of T-cells targeting a variety of

surface antigens, while engineered CAR vaccines typically target one key antigen. What

is your opinion about which strategy is best? Would the best approach vary from patient

to patient?

C. Some studies with TIL vaccines suggest that enriching and amplifying the T-cells from the

patient that are directed against a specific target antigen provide the best prognosis, while

other students suggest that using a mixture of TILs against a variety of antigens is most

successful. What is your opinion? How would we know which procedure to use in

advance?

4. Clinical Trial Patients:

A. Early cancer vaccine clinical trials have been criticized as having a relatively low number of

patients. Lately, the number of patients in cancer vaccine clinical trials has grown

exponentially to the point that some people worry there might not be a sufficient number

of refractory cancer patients to enroll in the trials. Do you think this is a problem?

5. Patient Chemoablation Treatments:

A. For TIL and CAR T-cell vaccines, some studies indicate that pre-treating the patient with

chemotherapy to obliterate (chemoablate) the patient’s own immune system prior to

Page 117: CANCER VACCINES - Worcester Polytechnic Institute

117

infusing the T-cell vaccine is critical for eliminating cells that can inhibit the vaccine.

Other studies indicate this pre-treatment is not needed. In your opinion, what dictates

when chemoablation should be used?

6. Immune Checkpoint Vaccines:

A. It appears that a recent trend in the cancer vaccine field has been to use combination vaccines

of 1) a checkpoint inhibitor plus 2) a vaccine against a specific target antigen. Do you

think this combination approach has been mostly successful?

B. What do you think of the recent failed Incyte Phase-III clinical trial using a PD-1 inhibitor

(Opdivo) plus an inhibitor of the enzyme IDO? Their phase-I and II data looked very

promising, but the phase-III data with the combination was no better than Opdivo alone.

What is your opinion?

C. Immune checkpoint vaccines have been shown in some cases to cause an over-activation of the

immune system (such as inflammation). Do you now routinely monitor for these effects?

7. Personalized Medicine:

A. Some scientists argue that cancer surface antigens vary from patient to patient, thus using a

personalized medicine approach to identify a patient’s own surface antigens is better than

using a vaccine that targets a generic surface antigen. But how expensive is this

personalized approach? It must be expensive to sequence the DNA from a patient’s

tumor to identify “neo-antigens” present on that particular tumor.

B. How long does a typical DNA genome (exome) sequencing project take? Our readings

indicate it can take weeks to sequence the DNA, analyze it for neo-antigen formation, and

synthesize the neo-antigens chemically. Some patients don’t have weeks to live, so

should the speed of this approach be accelerated?

INTERVIEW PREAMBLE

We are a group of students from the Worcester Polytechnic Institute in Massachusetts, and for our

research project we are conducting a series of interviews to investigate problems associated with the field

of cancer vaccines which have recently shown both strong successes and failures.

Your participation in this interview is completely voluntary, and you may withdraw at any time.

During this interview, we would like to record our conversation for later analysis. We will also be taking

notes during the interview on key points. Is this okay with you?

Can we also have your permission to quote any comments or perspectives expressed during the

interview? This information will be used for research purposes only, and we will give you an opportunity

to review any materials we use prior to the completion of our final report, which will be published on-line

in WPI’s archive of projects.

If the subject does not agree to be quoted, we will respond as follows: “Since you would not like

to be quoted during this interview, we will make sure your responses are anonymous. No names or

identifying information will appear in any of the project reports or publications.”

Your participation and assistance is greatly appreciated, and we thank you for taking the time to

meet with us. If you are interested, we would be happy to provide you with a copy of our results at the

conclusion of our study.